Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 19(8): 1122-1136, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39094561

RESUMO

Reactive astrocytes are known to exert detrimental effects upon neurons in several neurodegenerative diseases, yet our understanding of how astrocytes promote neurotoxicity remains incomplete, especially in human systems. In this study, we leveraged human pluripotent stem cell (hPSC) models to examine how reactivity alters astrocyte function and mediates neurodegeneration. hPSC-derived astrocytes were induced to a reactive phenotype, at which point they exhibited a hypertrophic profile and increased complement C3 expression. Functionally, reactive astrocytes displayed decreased intracellular calcium, elevated phagocytic capacity, and decreased contribution to the blood-brain barrier. Subsequently, co-culture of reactive astrocytes with a variety of neuronal cell types promoted morphological and functional alterations. Furthermore, when reactivity was induced in astrocytes from patient-specific hPSCs (glaucoma, Alzheimer's disease, and amyotrophic lateral sclerosis), the reactive state exacerbated astrocytic disease-associated phenotypes. These results demonstrate how reactive astrocytes modulate neurodegeneration, significantly contributing to our understanding of a role for reactive astrocytes in neurodegenerative diseases.


Assuntos
Astrócitos , Técnicas de Cocultura , Células-Tronco Pluripotentes , Astrócitos/metabolismo , Humanos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Complemento C3/metabolismo , Diferenciação Celular , Neurônios/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Fagocitose , Barreira Hematoencefálica/metabolismo , Glaucoma/patologia , Glaucoma/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Cálcio/metabolismo , Fenótipo
2.
Proc Natl Acad Sci U S A ; 121(25): e2317285121, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38870053

RESUMO

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.


Assuntos
Diferenciação Celular , Organoides , Células-Tronco Pluripotentes , Retina , Humanos , Organoides/citologia , Organoides/metabolismo , Retina/citologia , Retina/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Reprodutibilidade dos Testes , Proteínas Morfogenéticas Ósseas/metabolismo
3.
Sci Rep ; 13(1): 13827, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37620502

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of Aß 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 Aß42:Aß40 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.


Assuntos
Doença de Alzheimer , Humanos , Organoides , Sistema Nervoso Central , Fenótipo , Retina
4.
iScience ; 26(4): 106361, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37009209

RESUMO

Neuronal repopulation achieved through transplantation or transdifferentiation from endogenous sources holds tremendous potential for restoring function in chronic neurodegenerative disease or acute injury. Key to the evaluation of neuronal engraftment is the definitive discrimination of new or donor neurons from preexisting cells within the host tissue. Recent work has identified mechanisms by which genetically encoded donor cell reporters can be transferred to host neurons through intercellular material transfer. In addition, labeling transplanted and endogenously transdifferentiated neurons through viral vector transduction can yield misexpression in host cells in some circumstances. These issues can confound the tracking and evaluation of repopulated neurons in regenerative experimental paradigms. Using the retina as an example, we discuss common reasons for artifactual labeling of endogenous host neurons with donor cell reporters and suggest strategies to prevent erroneous conclusions based on misidentification of cell origin.

5.
Handb Exp Pharmacol ; 281: 83-102, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36907969

RESUMO

The ability to derive retinal ganglion cells (RGCs) from human pluripotent stem cells (hPSCs) provides an extraordinary opportunity to study the development of RGCs as well as cellular mechanisms underlying their degeneration in optic neuropathies. In the past several years, multiple approaches have been established that allow for the generation of RGCs from hPSCs, with these methods greatly improved in more recent studies to yield mature RGCs that more faithfully recapitulate phenotypes within the eye. Nevertheless, numerous differences still remain between hPSC-RGCs and those found within the human eye, with these differences likely explained at least in part due to the environment in which hPSC-RGCs are grown. With the ultimate goal of generating hPSC-RGCs that most closely resemble those within the retina for proper studies of retinal development, disease modeling, as well as cellular replacement, we review within this manuscript the current effective approaches for the differentiation of hPSC-RGCs, as well as how they have been applied for the investigation of RGC neurodegenerative diseases such as glaucoma. Furthermore, we provide our opinions on the characteristics of RGCs necessary for their use as effective in vitro disease models and importantly, how these current systems should be improved to more accurately reflect disease states. The establishment of characteristics in differentiated hPSC-RGCs that more effectively mimic RGCs within the retina will not only enable their use as effective models of RGC development, but will also create a better disease model for the identification of mechanisms underlying the neurodegeneration of RGCs in disease states such as glaucoma, further facilitating the development of therapeutic approaches to rescue RGCs from degeneration in disease states.


Assuntos
Glaucoma , Células-Tronco Pluripotentes , Humanos , Células Ganglionares da Retina , Retina , Diferenciação Celular , Glaucoma/terapia
6.
Commun Biol ; 6(1): 218, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36828933

RESUMO

Mitochondrial dysfunctions are widely afflicted in central nervous system (CNS) disorders with minimal understanding on how to improve mitochondrial homeostasis to promote neuroprotection. Here we have used human stem cell differentiated retinal ganglion cells (hRGCs) of the CNS, which are highly sensitive towards mitochondrial dysfunctions due to their unique structure and function, to identify mechanisms for improving mitochondrial quality control (MQC). We show that hRGCs are efficient in maintaining mitochondrial homeostasis through rapid degradation and biogenesis of mitochondria under acute damage. Using a glaucomatous Optineurin mutant (E50K) stem cell line, we show that at basal level mutant hRGCs possess less mitochondrial mass and suffer mitochondrial swelling due to excess ATP production load. Activation of mitochondrial biogenesis through pharmacological inhibition of the Tank binding kinase 1 (TBK1) restores energy homeostasis, mitigates mitochondrial swelling with neuroprotection against acute mitochondrial damage for glaucomatous E50K hRGCs, revealing a novel neuroprotection mechanism.


Assuntos
Glaucoma , Células-Tronco Pluripotentes , Humanos , Células Ganglionares da Retina , Biogênese de Organelas , Neuroproteção , Mitocôndrias/metabolismo , Células-Tronco Pluripotentes/metabolismo
7.
bioRxiv ; 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36711831

RESUMO

Autophagy dysfunction has been associated with several neurodegenerative diseases including glaucoma, characterized by the degeneration of retinal ganglion cells (RGCs). However, the mechanisms by which autophagy dysfunction promotes RGC damage remain unclear. Here, we hypothesized that perturbation of the autophagy pathway results in increased autophagic demand, thereby downregulating signaling through mammalian target of rapamycin complex 1 (mTORC1), a negative regulator of autophagy, contributing to the degeneration of RGCs. We identified an impairment of autophagic-lysosomal degradation and decreased mTORC1 signaling via activation of the stress sensor adenosine monophosphate-activated protein kinase (AMPK), along with subsequent neurodegeneration in RGCs differentiated from human pluripotent stem cells (hPSCs) with a glaucoma-associated variant of Optineurin (OPTN-E50K). Similarly, the microbead occlusion model of glaucoma resulting in ocular hypertension also exhibited autophagy disruption and mTORC1 downregulation. Pharmacological inhibition of mTORC1 in hPSC-derived RGCs recapitulated disease-related neurodegenerative phenotypes in otherwise healthy RGCs, while the mTOR-independent induction of autophagy reduced protein accumulation and restored neurite outgrowth in diseased OPTN-E50K RGCs. Taken together, these results highlight an important balance between autophagy and mTORC1 signaling essential for RGC homeostasis, while disruption to these pathways contributes to neurodegenerative features in glaucoma, providing a potential therapeutic target to prevent neurodegeneration.

8.
Stem Cell Reports ; 17(7): 1636-1649, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35714595

RESUMO

Although the degeneration of retinal ganglion cells (RGCs) is a primary characteristic of glaucoma, astrocytes also contribute to their neurodegeneration in disease states. Although studies often explore cell-autonomous aspects of RGC neurodegeneration, a more comprehensive model of glaucoma should take into consideration interactions between astrocytes and RGCs. To explore this concept, RGCs and astrocytes were differentiated from human pluripotent stem cells (hPSCs) with a glaucoma-associated OPTN(E50K) mutation along with corresponding isogenic controls. Initial results indicated significant changes in OPTN(E50K) astrocytes, including evidence of autophagy dysfunction. Subsequently, co-culture experiments demonstrated that OPTN(E50K) astrocytes led to neurodegenerative properties in otherwise healthy RGCs, while healthy astrocytes rescued some neurodegenerative features in OPTN(E50K) RGCs. These results are the first to identify disease phenotypes in OPTN(E50K) astrocytes, including how their modulation of RGCs is affected. Moreover, these results support the concept that astrocytes could offer a promising target for therapeutic intervention in glaucoma.


Assuntos
Glaucoma , Células-Tronco Pluripotentes , Astrócitos , Proteínas de Ciclo Celular/genética , Glaucoma/genética , Humanos , Proteínas de Membrana Transportadoras/genética , Fenótipo , Células Ganglionares da Retina
9.
Stem Cell Reports ; 16(9): 2228-2241, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34115986

RESUMO

The development of the visual system involves the coordination of spatial and temporal events to specify the organization of varied cell types, including the elongation of axons from retinal ganglion cells (RGCs) to post-synaptic targets in the brain. Retinal organoids recapitulate many features of retinal development, yet have lacked downstream targets into which RGC axons extend, limiting the ability to model projections of the human visual system. To address these issues, retinal organoids were generated and organized into an in vitro assembloid model of the visual system with cortical and thalamic organoids. RGCs responded to environmental cues and extended axons deep into assembloids, modeling the projections of the visual system. In addition, RGC survival was enhanced in long-term assembloids, overcoming prior limitations of retinal organoids in which RGCs are lost. Overall, these approaches will facilitate studies of human visual system development, as well as diseases or injuries to this critical pathway.


Assuntos
Diferenciação Celular , Organoides/citologia , Organoides/metabolismo , Células-Tronco Pluripotentes/citologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/fisiologia , Animais , Axônios/fisiologia , Biomarcadores , Técnicas de Cultura de Células em Três Dimensões/métodos , Fenômenos Fisiológicos Celulares , Células Cultivadas , Imunofluorescência , Genes Reporter , Humanos , Camundongos , Crescimento Neuronal , Sinapses/metabolismo , Vias Visuais
10.
Sci Rep ; 10(1): 17359, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060618

RESUMO

Retinal ganglion cells (RGCs) are a heterogeneous population of neurons, comprised of numerous subtypes that work synchronously to transmit visual information to the brain. In blinding disorders such as glaucoma, RGCs are the main cell type to degenerate and lead to loss of vision. Previous studies have identified and characterized a variety of RGC subtypes in animal models, although only a handful of studies demonstrate the differential loss of these RGC subtypes in response to disease or injury. Thus, efforts of the current study utilized both chronic (bead occlusion) and acute (optic nerve crush, ONC) rat models to characterize disease response and differential loss of RGC subtypes. Bead occlusion and ONC retinas demonstrated significant RGC loss, glial reactivity and apoptosis compared to control retinas. Importantly, bead occlusion and ONC retinas resulted in differential subtype-specific loss of RGCs, with a high susceptibility for alpha- and direction selective-RGCs and preferential survival of ipRGCs. Results of this study serve as an important foundation for future experiments focused on the mechanisms resulting in the loss of RGCs in optic neuropathies, as well as the development of targeted therapeutics for RGC subtype-specific neuroprotection.


Assuntos
Modelos Animais de Doenças , Modelos Biológicos , Células Ganglionares da Retina/patologia , Doença Aguda , Animais , Doença Crônica , Fármacos Neuroprotetores/farmacologia , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/prevenção & controle , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Células Ganglionares da Retina/efeitos dos fármacos
11.
Stem Cell Reports ; 15(1): 52-66, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32531194

RESUMO

Retinal ganglion cells (RGCs) serve as the connection between the eye and the brain, with this connection disrupted in glaucoma. Numerous cellular mechanisms have been associated with glaucomatous neurodegeneration, and useful cellular models of glaucoma allow for the precise analysis of degenerative phenotypes. Human pluripotent stem cells (hPSCs) serve as powerful tools for studying human disease, particularly cellular mechanisms underlying neurodegeneration. Thus, efforts focused upon hPSCs with an E50K mutation in the Optineurin (OPTN) gene, a leading cause of inherited forms of glaucoma. CRISPR/Cas9 gene editing introduced the OPTN(E50K) mutation into existing lines of hPSCs, as well as generating isogenic controls from patient-derived lines. RGCs differentiated from OPTN(E50K) hPSCs exhibited numerous neurodegenerative deficits, including neurite retraction, autophagy dysfunction, apoptosis, and increased excitability. These results demonstrate the utility of OPTN(E50K) RGCs as an in vitro model of neurodegeneration, with the opportunity to develop novel therapeutic approaches for glaucoma.


Assuntos
Proteínas de Ciclo Celular/genética , Glaucoma/genética , Proteínas de Membrana Transportadoras/genética , Mutação/genética , Degeneração Neural/patologia , Organoides/patologia , Células Ganglionares da Retina/patologia , Animais , Apoptose , Autofagia , Sistemas CRISPR-Cas/genética , Diferenciação Celular/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Edição de Genes , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Fenótipo , Análise de Sequência de RNA
12.
Methods Cell Biol ; 159: 279-302, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32586447

RESUMO

Human pluripotent stem cells (hPSCs) possess the remarkable ability to differentiate into any cell type of the body, including those of the retina. Through the differentiation of these cells as retinal organoids, it is now possible to model the spatial and temporal development of the human retina using hPSCs, in which retinal progenitor cells produce the entire repertoire of retinal cells, first differentiating into retinal ganglion cells and ending with mature photoreceptors, bipolar cells, and Müller glia. Importantly, retinal organoids self-assemble into laminated structures that recapitulate the layering of the human retina with a retinal ganglion cell layer lining the inner layer and a distinctly separate photoreceptor layer occupying the outer layers. This organoid technology has provided access to human tissue for developmental and disease modeling, as well as translational applications such as high throughput drug screening and cell replacement therapies. However, the differentiation of retinal organoids does require some expertise and multiple strategies produce inconsistent results. Here, we describe in detail a well-established and relatively simple method for the generation of retinal organoids.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Agregação Celular , Humanos
13.
Prog Neurobiol ; 193: 101817, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32360241

RESUMO

Glaucoma, one of the most common causes of blindness in developing countries today, involves a progressive loss of neural cells in the optic nerve that leads to progressive, irreversible vision loss. Increased intraocular pressure (IOP) presents as a major risk factor for glaucoma, although there exist cases of glaucoma patients with normal IOP that exhibit damage to retinal ganglion cells (RGCs) and the optic nerve. However, treatment approaches have maintained their focus on modifying IOP due to a lack of other modifiable risks factors. Traditional concepts in glaucoma involve the neuronal environment and external effects as a source of causative factors; however, studies have yet to investigate whether the molecular profile of RGCs in glaucoma patients makes them more vulnerable and/or susceptible to external damage. Our hypothesis states that molecular changes at the whole cell, gene expression, and electrophysiological level of the neurons can contribute to their degeneration. Herein, we briefly describe different types of glaucoma and any similarities to different molecular and cellular features of neurodegeneration. To test our hypothesis, we describe human induced pluripotent stem cells (hiPSCs) as a reliable cellular tool to model neurodegenerative aspects of glaucoma to reveal the multiple pathological molecular mechanisms underlying disease development.


Assuntos
Predisposição Genética para Doença , Glaucoma , Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Células Ganglionares da Retina , Glaucoma/etiologia , Glaucoma/genética , Glaucoma/metabolismo , Glaucoma/patologia , Humanos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia
14.
J Neurosci ; 40(20): 3896-3914, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32300046

RESUMO

Optic neuropathies are a group of optic nerve (ON) diseases caused by various insults including glaucoma, inflammation, ischemia, trauma, and genetic deficits, which are characterized by retinal ganglion cell (RGC) death and ON degeneration. An increasing number of genes involved in RGC intrinsic signaling have been found to be promising neural repair targets that can potentially be modulated directly by gene therapy, if we can achieve RGC specific gene targeting. To address this challenge, we first used adeno-associated virus (AAV)-mediated gene transfer to perform a low-throughput in vivo screening in both male and female mouse eyes and identified the mouse γ-synuclein (mSncg) promoter, which specifically and potently sustained transgene expression in mouse RGCs and also works in human RGCs. We further demonstrated that gene therapy that combines AAV-mSncg promoter with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing can knock down pro-degenerative genes in RGCs and provide effective neuroprotection in optic neuropathies.SIGNIFICANCE STATEMENT Here, we present an RGC-specific promoter, mouse γ-synuclein (mSncg) promoter, and perform extensive characterization and proof-of-concept studies of mSncg promoter-mediated gene expression and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing in RGCs in vivo To our knowledge, this is the first report demonstrating in vivo neuroprotection of injured RGCs and optic nerve (ON) by AAV-mediated CRISPR/Cas9 inhibition of genes that are critical for neurodegeneration. It represents a powerful tool to achieve RGC-specific gene modulation, and also opens up a promising gene therapy strategy for optic neuropathies, the most common form of eye diseases that cause irreversible blindness.


Assuntos
Regulação da Expressão Gênica/genética , Edição de RNA/genética , Células Ganglionares da Retina/metabolismo , gama-Sinucleína/genética , Animais , Sistemas CRISPR-Cas , Dependovirus/genética , Feminino , Deleção de Genes , Terapia Genética , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nervo Óptico/patologia , Doenças do Nervo Óptico/genética , Doenças do Nervo Óptico/terapia , Células Ganglionares da Retina/patologia , Transgenes/genética
15.
Mol Neurobiol ; 57(2): 668-684, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31435904

RESUMO

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.


Assuntos
Corpo Estriado/metabolismo , Metabolismo Energético , Proteína Huntingtina/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/metabolismo , Proteínas Mutantes/metabolismo , Neurônios/metabolismo , Superóxidos/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Astrócitos/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Potencial da Membrana Mitocondrial , Expansão das Repetições de Trinucleotídeos/genética
16.
Adv Exp Med Biol ; 1186: 121-140, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31654388

RESUMO

Human pluripotent stem cell (hPSC) technology has revolutionized the field of biology through the unprecedented ability to study the differentiation of human cells in vitro. In the past decade, hPSCs have been applied to study development, model disease, develop drugs, and devise cell replacement therapies for numerous biological systems. Of particular interest is the application of this technology to study and treat optic neuropathies such as glaucoma. Retinal ganglion cells (RGCs) are the primary cell type affected in these diseases, and once lost, they are unable to regenerate in adulthood. This necessitates the development of strategies to study the mechanisms of degeneration as well as develop translational therapeutic approaches to treat early- and late-stage disease progression. Numerous protocols have been established to derive RGCs from hPSCs, with the ability to generate large populations of human RGCs for translational applications. In this review, the key applications of hPSCs within the retinal field are described, including the use of these cells as developmental models, disease models, drug development, and finally, cell replacement therapies. In greater detail, the current report focuses on the differentiation of hPSC-derived RGCs and the many unique characteristics associated with these cells in vitro including their genetic identifiers, their electrophysiological activity, and their morphological maturation. Also described is the current progress in the use of patient-specific hPSCs to study optic neuropathies affecting RGCs, with emphasis on the use of these RGCs for studying disease mechanisms and pathogenesis, drug screening, and cell replacement therapies in future studies.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes , Células Ganglionares da Retina , Glaucoma/terapia , Humanos , Neuropatologia/tendências , Doenças do Nervo Óptico/terapia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Retina/patologia , Células Ganglionares da Retina/citologia
17.
Stem Cell Reports ; 12(2): 201-212, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30639213

RESUMO

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.


Assuntos
Astrócitos/citologia , Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Células Ganglionares da Retina/citologia , Células Cultivadas , Humanos
18.
Sci Rep ; 8(1): 14520, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30266927

RESUMO

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.


Assuntos
Organoides/citologia , Células-Tronco Pluripotentes/citologia , Células Ganglionares da Retina/citologia , Meios de Cultura , Genes Reporter , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Crescimento Neuronal , Proteína Vermelha Fluorescente
19.
Stem Cell Reports ; 10(4): 1282-1293, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29576537

RESUMO

Retinal ganglion cells (RGCs) are the projection neurons of the retina and transmit visual information to postsynaptic targets in the brain. While this function is shared among nearly all RGCs, this class of cell is remarkably diverse, comprised of multiple subtypes. Previous efforts have identified numerous RGC subtypes in animal models, but less attention has been paid to human RGCs. Thus, efforts of this study examined the diversity of RGCs differentiated from human pluripotent stem cells (hPSCs) and characterized defined subtypes through the expression of subtype-specific markers. Further investigation of these subtypes was achieved using single-cell transcriptomics, confirming the combinatorial expression of molecular markers associated with these subtypes, and also provided insight into more subtype-specific markers. Thus, the results of this study describe the derivation of RGC subtypes from hPSCs and will support the future exploration of phenotypic and functional diversity within human RGCs.


Assuntos
Células-Tronco Pluripotentes/citologia , Células Ganglionares da Retina/citologia , Biomarcadores/metabolismo , Forma Celular , Células Cultivadas , Proteínas do Domínio Duplacortina , Regulação da Expressão Gênica , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Transcrição Gênica
20.
Stem Cells Int ; 2016: 3241057, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27642304
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA