Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Front Genet ; 14: 1251216, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37745862

RESUMO

Dominant optic atrophy (DOA) is an inherited disease that leads to the loss of retinal ganglion cells (RGCs), the projection neurons that relay visual information from the retina to the brain through the optic nerve. The majority of DOA cases can be attributed to mutations in optic atrophy 1 (OPA1), a nuclear gene encoding a mitochondrial-targeted protein that plays important roles in maintaining mitochondrial structure, dynamics, and bioenergetics. Although OPA1 is ubiquitously expressed in all human tissues, RGCs appear to be the primary cell type affected by OPA1 mutations. DOA has not been extensively studied in human RGCs due to the general unavailability of retinal tissues. However, recent advances in stem cell biology have made it possible to produce human RGCs from pluripotent stem cells (PSCs). To aid in establishing DOA disease models based on human PSC-derived RGCs, we have generated iPSC lines from two DOA patients who carry distinct OPA1 mutations and present very different disease symptoms. Studies using these OPA1 mutant RGCs can be correlated with clinical features in the patients to provide insights into DOA disease mechanisms.

2.
Nat Commun ; 13(1): 7037, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396639

RESUMO

Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective cytokine in multiple models of retinal degeneration. To understand mechanisms underlying its broad neuroprotective effects, we have investigated the influence of CNTF on metabolism in a mouse model of photoreceptor degeneration. CNTF treatment improves the morphology of photoreceptor mitochondria, but also leads to reduced oxygen consumption and suppressed respiratory chain activities. Molecular analyses show elevated glycolytic pathway gene transcripts and active enzymes. Metabolomics analyses detect significantly higher levels of ATP and the energy currency phosphocreatine, elevated glycolytic pathway metabolites, increased TCA cycle metabolites, lipid biosynthetic pathway intermediates, nucleotides, and amino acids. Moreover, CNTF treatment restores the key antioxidant glutathione to the wild type level. Therefore, CNTF significantly impacts the metabolic status of degenerating retinas by promoting aerobic glycolysis and augmenting anabolic activities. These findings reveal cellular mechanisms underlying enhanced neuronal viability and suggest potential therapies for treating retinal degeneration.


Assuntos
Fator Neurotrófico Ciliar , Degeneração Retiniana , Camundongos , Animais , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Degeneração Retiniana/terapia , Neuroproteção , Retina/metabolismo , Glicólise
3.
Stem Cell Res ; 56: 102516, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34469777

RESUMO

The homozygous autosomal recessive truncating mutations of LDL receptor related protein associated protein 1 (LRPAP1) is a possible reason for Nonsyndromic Extreme Myopia, patients with which show typical chorioretinal degeneration. We generated an LRPAP1 knockout FDCHDPe009-B embryonic stem cell line to study mechanisms of retinal degeneration underlying LRPAP1 deficiency with the help of the CRISPR/Cas9 system. Two distinct biallelic deletions in the cell line have been confirmed, which causing a frameshift and premature stop codons thus influence the translation of LRPAP1. FDCHDPe009-B has maintained normal stem cell morphology, pluripotent gene expression, parental karyotype, and ability to differentiate into three germ layers.


Assuntos
Sistemas CRISPR-Cas , Células-Tronco Embrionárias Humanas , Sistemas CRISPR-Cas/genética , Linhagem Celular , Células-Tronco Embrionárias , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade
4.
Front Cell Dev Biol ; 9: 653305, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34055784

RESUMO

The developing retina expresses multiple bHLH transcription factors. Their precise functions and interactions in uncommitted retinal progenitors remain to be fully elucidated. Here, we investigate the roles of bHLH factors ATOH7 and Neurog2 in human ES cell-derived retinal organoids. Single cell transcriptome analyses identify three states of proliferating retinal progenitors: pre-neurogenic, neurogenic, and cell cycle-exiting progenitors. Each shows different expression profile of bHLH factors. The cell cycle-exiting progenitors feed into a postmitotic heterozygous neuroblast pool that gives rise to early born neuronal lineages. Elevating ATOH7 or Neurog2 expression accelerates the transition from the pre-neurogenic to the neurogenic state, and expands the exiting progenitor and neuroblast populations. In addition, ATOH7 and Neurog2 significantly, yet differentially, enhance retinal ganglion cell and cone photoreceptor production. Moreover, single cell transcriptome analyses reveal that ATOH7 and Neurog2 each assert positive autoregulation, and both suppress key bHLH factors associated with the pre-neurogenic and states and elevate bHLH factors expressed by exiting progenitors and differentiating neuroblasts. This study thus provides novel insight regarding how ATOH7 and Neurog2 impact human retinal progenitor behaviors and neuroblast fate choices.

5.
Stem Cell Res ; 53: 102342, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33878707

RESUMO

LRP2 is mainly expressed in the cell membrane of epithelia, maintaining normal endocytosis of nutrients from the extracellular microenvironment and mediating growth factor signals. The deficiency of LRP2 can result in abnormal lysosomal and mitochondrial function as well as insufficient resistance to oxidative stress. LRP2-KO animals show enlarged eyes and malfunction of the retinal pigment epithelium (RPE). We were able to generate an LRP2-KO human embryonic stem (ES) cell line using CRISPR/Cas9 gene editing and differentiate the mutant ES cells into RPE cells. Thus, this LRP2-KO human ES line will facilitate studying cellular mechanisms of eye disease due to LRP2 deficiency.


Assuntos
Sistemas CRISPR-Cas , Células-Tronco Embrionárias Humanas , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo
6.
Sci Rep ; 10(1): 6593, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313077

RESUMO

Ciliary neurotrophic factor (CNTF) has been tested in clinical trials for human retinal degeneration due to its potent neuroprotective effects in various animal models. To decipher CNTF-triggered molecular events in the degenerating retina, we performed high-throughput RNA sequencing analyses using the Rds/Prph2 (P216L) transgenic mouse as a preclinical model for retinitis pigmentosa. In the absence of CNTF treatment, transcriptome alterations were detected at the onset of rod degeneration compared with wild type mice, including reduction of key photoreceptor transcription factors Crx, Nrl, and rod phototransduction genes. Short-term CNTF treatments caused further declines of photoreceptor transcription factors accompanied by marked decreases of both rod- and cone-specific gene expression. In addition, CNTF triggered acute elevation of transcripts in the innate immune system and growth factor signaling. These immune responses were sustained after long-term CNTF exposures that also affected neuronal transmission and metabolism. Comparisons of transcriptomes also uncovered common pathways shared with other retinal degeneration models. Cross referencing bulk RNA-seq with single-cell RNA-seq data revealed the CNTF responsive cell types, including Müller glia, rod and cone photoreceptors, and bipolar cells. Together, these results demonstrate the influence of exogenous CNTF on the retinal transcriptome landscape and illuminate likely CNTF impacts in degenerating human retinas.


Assuntos
Fator Neurotrófico Ciliar/farmacologia , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/genética , Transcriptoma/genética , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Ratos , Retina/efeitos dos fármacos , Degeneração Retiniana/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
7.
Stem Cell Reports ; 13(4): 747-760, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31543471

RESUMO

The development of the mammalian retina is a complicated process involving the generation of distinct types of neurons from retinal progenitor cells (RPCs) in a spatiotemporal-specific manner. The progression of RPCs during retinogenesis includes RPC proliferation, cell-fate commitment, and specific neuronal differentiation. In this study, by performing single-cell RNA sequencing of cells isolated from human embryonic stem cell (hESC)-derived 3D retinal organoids, we successfully deconstructed the temporal progression of RPCs during early human retinogenesis. We identified two distinctive subtypes of RPCs with unique molecular profiles, namely multipotent RPCs and neurogenic RPCs. We found that genes related to the Notch and Wnt signaling pathways, as well as chromatin remodeling, were dynamically regulated during RPC commitment. Interestingly, our analysis identified that CCND1, a G1-phase cell-cycle regulator, was coexpressed with ASCL1 in a cell-cycle-independent manner. Temporally controlled overexpression of CCND1 in retinal organoids demonstrated a role for CCND1 in promoting early retinal neurogenesis. Together, our results revealed critical pathways and novel genes in early retinogenesis of humans.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Organogênese/genética , Organoides , Retina/citologia , Retina/metabolismo , Biomarcadores , Imunofluorescência , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunofenotipagem , Análise de Célula Única , Técnicas de Cultura de Tecidos
8.
Stem Cell Res ; 39: 101495, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31376721

RESUMO

Pathological myopia (PM) is a major cause of irreversible vision impairment worldwide. We have successfully reprogrammed the peripheral blood mononuclear cells (PBMCs) from a PM patient to induced pluripotent stem cells and characterized their pluripotency and genetic stability, as well as the potential to differentiate to retinal pigment epithelium (RPE). This line may serve as a useful tool to explore the pathogenesis of PM.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Miopia Degenerativa/terapia , Epitélio Pigmentado da Retina/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Humanos , Imuno-Histoquímica , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/fisiologia
9.
Stem Cell Res ; 38: 101459, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31146250

RESUMO

Best's disease (BD) is an inherited retinal degenerative disease caused by mutations in BEST1 gene. A human induced pluripotent stem cell (iPSC) line has been generated with integration-free Sendai virus method from peripheral blood mononuclear cells (PBMCs) of a BD patient carrying c.888C > A mutation in BEST1 gene. This cell line may serve as a model for the study of pathogenesis of BD.


Assuntos
Bestrofinas , Células-Tronco Pluripotentes Induzidas , Mutação de Sentido Incorreto , Distrofia Macular Viteliforme , Substituição de Aminoácidos , Bestrofinas/genética , Bestrofinas/metabolismo , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo , Distrofia Macular Viteliforme/patologia
10.
Sci Rep ; 8(1): 6823, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717171

RESUMO

The production of vertebrate retinal projection neurons, retinal ganglion cells (RGCs), is regulated by cell-intrinsic determinants and cell-to-cell signaling events. The basic-helix-loop-helix (bHLH) protein Atoh7 is a key neurogenic transcription factor required for RGC development. Here, we investigate whether manipulating human ATOH7 expression among uncommitted progenitors can promote RGC fate specification and thus be used as a strategy to enhance RGC genesis. Using the chicken retina as a model, we show that cell autonomous expression of ATOH7 is sufficient to induce precocious RGC formation and expansion of the neurogenic territory. ATOH7 overexpression among neurogenic progenitors significantly enhances RGC production at the expense of reducing the progenitor pool. Furthermore, forced expression of ATOH7 leads to a minor increase of cone photoreceptors. We provide evidence that elevating ATOH7 levels accelerates cell cycle progression from S to M phase and promotes cell cycle exit. We also show that ATOH7-induced ectopic RGCs often exhibit aberrant axonal projection patterns and are correlated with increased cell death during the period of retinotectal connections. These results demonstrate the high potency of human ATOH7 in promoting early retinogenesis and specifying the RGC differentiation program, thus providing insight for manipulating RGC production from stem cell-derived retinal organoids.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Células Ganglionares da Retina/fisiologia , Células-Tronco/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Embrião de Galinha , Vetores Genéticos , Humanos , Modelos Animais , Neurogênese/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Retroviridae/genética
11.
Dev Dyn ; 245(7): 727-38, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26661417

RESUMO

Retinal dystrophies are a major cause of blindness for which there are currently no curative treatments. Transplantation of stem cell-derived neuronal progenitors to replace lost cells has been widely investigated as a therapeutic option. Another promising strategy would be to trigger self-repair mechanisms in patients, through the recruitment of endogenous cells with stemness properties. Accumulating evidence in the past 15 year0s has revealed that several retinal cell types possess neurogenic potential, thus opening new avenues for regenerative medicine. Among them, Müller glial cells have been shown to be able to undergo a reprogramming process to re-acquire a stem/progenitor state, allowing them to proliferate and generate new neurons for repair following retinal damages. Although Müller cell-dependent spontaneous regeneration is remarkable in some species such as the fish, it is extremely limited and ineffective in mammals. Understanding the cellular events and molecular mechanisms underlying Müller cell activities in species endowed with regenerative capacities could provide knowledge to unlock the restricted potential of their mammalian counterparts. In this context, the present review provides an overview of Müller cell responses to injury across vertebrate model systems and summarizes recent advances in this rapidly evolving field. Developmental Dynamics 245:727-738, 2016. © 2015 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc.


Assuntos
Células Ependimogliais/citologia , Células Ependimogliais/fisiologia , Retina/citologia , Animais , Células Ependimogliais/metabolismo , Humanos , Regeneração/genética , Regeneração/fisiologia , Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
12.
PLoS One ; 9(11): e112175, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25401462

RESUMO

The neural retina is a critical component of the visual system, which provides the majority of sensory input in humans. Various retinal degenerative diseases can result in the permanent loss of retinal neurons, especially the light-sensing photoreceptors and the centrally projecting retinal ganglion cells (RGCs). The replenishment of lost RGCs and the repair of optic nerve damage are particularly challenging, as both RGC specification and their subsequent axonal growth and projection involve complex and precise regulation. To explore the developmental potential of pluripotent stem cell-derived neural progenitors, we have established mouse iPS cells that allow cell lineage tracing of progenitors that have expressed Atoh7/Math5, a bHLH transcription factor required for RGC production. These Atoh7 lineage reporter iPS cells encode Cre to replace one copy of the endogenous Atoh7 gene and a Cre-dependent YFP reporter in the ROSA locus. In addition, they express pluripotent markers and are capable of generating teratomas in vivo. Under anterior neural induction and neurogenic conditions in vitro, the Atoh7-Cre/ROSA-YFP iPS cells differentiate into neurons that co-express various RGC markers and YFP, indicating that these neurons are derived from Atoh7-expressing progenitors. Consistent with previous in vivo cell lineage studies, the Atoh7-Cre/ROSA-YFP iPS cells also give rise to a subset of Crx-positive photoreceptor precursors. Furthermore, inhibition of Notch signaling in the iPSC cultures results in a significant increase of YFP-positive RGCs and photoreceptor precursors. Together, these results show that Atoh7-Cre/ROSA-YFP iPS cells can be used to monitor the development and survival of RGCs and photoreceptors from pluripotent stem cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas do Tecido Nervoso/genética , Células Fotorreceptoras/citologia , Células Ganglionares da Retina/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores , Expressão Gênica , Genes Reporter , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Células Fotorreceptoras/metabolismo , Células Ganglionares da Retina/metabolismo
13.
Opt Express ; 22(16): 19610-20, 2014 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-25321044

RESUMO

Based on the framework of spin-flip model (SFM), the output characteristics of a 1550 nm vertical-cavity surface-emitting laser (VCSEL) subject to variable-polarization fiber Bragg grating (FBG) feedback (VPFBGF) have been investigated. With the aid of the self-correlation function (SF) and the permutation entropy (PE) function, the time-delay signature (TDS) of chaos in the VPFBGF-VCSEL is evaluated, and then the influences of the operation parameters on the TDS of chaos are analyzed. The results show that the TDS of chaos can be suppressed efficiently through selecting suitable coupling coefficient and feedback rate of the FBG, and is weaker than that of chaos generated by traditional variable-polarization mirror feedback VCSELs (VPMF-VCSELs) or polarization-preserved FBG feedback VCSELs (PPFBGF-VCSELs).

14.
Proc Natl Acad Sci U S A ; 110(47): E4520-9, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24191003

RESUMO

Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective agent in multiple retinal degeneration animal models. Recently, CNTF has been evaluated in clinical trials for the inherited degenerative disease retinitis pigmentosa (RP) and for dry age-related macular degeneration (AMD). Despite its potential as a broad-spectrum therapeutic treatment for blinding diseases, the target cells of exogenous CNTF and its mechanism of action remain poorly understood. We have shown previously that constitutive expression of CNTF prevents photoreceptor death but alters the retinal transcriptome and suppresses visual function. Here, we use a lentivirus to deliver the same secreted human CNTF used in clinical trials to a mouse model of RP. We found that low levels of CNTF halt photoreceptor death, improve photoreceptor morphology, and correct opsin mislocalization. However, we did not detect corresponding improvement of retinal function as measured by the electroretinogram. Disruption of the cytokine receptor gp130 gene in Müller glia reduces CNTF-dependent photoreceptor survival and prevents phosphorylation of STAT3 and ERK in Müller glia and the rest of the retina. Targeted deletion of gp130 in rods also demolishes neuroprotection by CNTF and prevents further activation of Müller glia. Moreover, CNTF elevates the expression of LIF and endothelin 2, thus positively promoting Müller and photoreceptor interactions. We propose that exogenous CNTF initially targets Müller glia, and subsequently induces cytokines acting through gp130 in photoreceptors to promote neuronal survival. These results elucidate a cellular mechanism for exogenous CNTF-triggered neuroprotection and provide insight into the complex cellular responses induced by CNTF in diseased retinas.


Assuntos
Fator Neurotrófico Ciliar/metabolismo , Receptor gp130 de Citocina/metabolismo , Células Ependimogliais/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneração Retiniana/tratamento farmacológico , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Lentivirus , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real , Degeneração Retiniana/genética
15.
Methods Mol Biol ; 884: 183-92, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22688706

RESUMO

Primary neuronal culture and transfection are useful tools in determining gene functions within specific tissue contexts and developmental stages. Chicken embryonic retinal cultures are easily obtainable and often robust as the chicken eye is relatively large compared to mouse eye at similar developmental stages. Various DNA-based constructs have been developed to overexpress or knockdown genes of interest and can be delivered into the cells using lipofectamine, a cationic lipid-based transfection system. Here, we describe a method to culture and transfect primary chicken embryonic retinal cells in order to manipulate genes involved in retinal development. This technique can simultaneously deliver multiple genes without construct-size constrains and permit the usage of tissue or cell type-specific promoters, and is thus a useful approach to explore gene functions during neural retina differentiation.


Assuntos
Lipídeos , Retina/metabolismo , Transfecção , Animais , Embrião de Galinha , DNA/metabolismo , Imuno-Histoquímica , Retina/citologia , Técnicas de Cultura de Tecidos
16.
Mol Cell Neurosci ; 49(2): 171-83, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22155156

RESUMO

The lipid phosphatase PTEN is a critical negative regulator of extracellular signal-induced PI3K activities, yet the roles of PTEN in the neural retina remain poorly understood. Here, we investigate the function of PTEN during retinal development. Deletion of Pten at the onset of neurogenesis in retinal progenitors results in the reduction of retinal ganglion cells and rod photoreceptors, but increased Müller glial genesis. In addition, PTEN deficiency leads to elevated phosphorylation of Akt, especially in the developing inner plexiform layer, where high levels of PTEN are normally expressed. In Pten mutant retinas, various subtypes of amacrine cells show severe dendritic overgrowth, causing specific expansion of the inner plexiform layer. However, the outer plexiform layer remains relatively undisturbed in the Pten deficient retina. Physiological analysis detects reduced rod function and augmented oscillatory potentials originating from amacrine cells in Pten mutants. Furthermore, deleting Pten or elevating Akt activity in individual amacrine cells is sufficient to disrupt dendritic arborization, indicating that Pten activity is required cell autonomously to control neuronal morphology. Moreover, inhibiting endogenous Akt activity attenuates inner plexiform layer formation in vitro. Together, these findings demonstrate that suppression of PI3K/Akt signaling by PTEN is crucial for proper neuronal differentiation and normal retinal network formation.


Assuntos
Interneurônios/fisiologia , PTEN Fosfo-Hidrolase/fisiologia , Retina/citologia , Células Amácrinas/citologia , Animais , Técnicas de Inativação de Genes , Interneurônios/citologia , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/genética , Morfogênese/fisiologia , Mutação/genética , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/classificação , Células Ganglionares da Retina/citologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Hum Mol Genet ; 19(21): 4229-38, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20709808

RESUMO

Age-related macular degeneration (AMD) is characterized by the loss or dysfunction of retinal pigment epithelium (RPE) and is the most common cause of vision loss among the elderly. Stem-cell-based strategies, using human embryonic stem cells (hESCs) or human-induced pluripotent stem cells (hiPSCs), may provide an abundant donor source for generating RPE cells in cell replacement therapies. Despite a significant amount of research on deriving functional RPE cells from various stem cell sources, it is still unclear whether stem-cell-derived RPE cells fully mimic primary RPE cells. In this report, we demonstrate that functional RPE cells can be derived from multiple lines of hESCs and hiPSCs with varying efficiencies. Stem-cell-derived RPE cells exhibit cobblestone-like morphology, transcripts, proteins and phagocytic function similar to human fetal RPE (fRPE) cells. In addition, we performed global gene expression profiling of stem-cell-derived RPE cells, native and cultured fRPE cells, undifferentiated hESCs and fibroblasts to determine the differentiation state of stem-cell-derived RPE cells. Our data indicate that hESC-derived RPE cells closely resemble human fRPE cells, whereas hiPSC-derived RPE cells are in a unique differentiation state. Furthermore, we identified a set of 87 signature genes that are unique to human fRPE and a majority of these signature genes are shared by stem-cell-derived RPE cells. These results establish a panel of molecular markers for evaluating the fidelity of human pluripotent stem cell to RPE conversion. This study contributes to our understanding of the utility of hESC/hiPSC-derived RPE in AMD therapy.


Assuntos
Epitélio Pigmentado da Retina/metabolismo , Células-Tronco/metabolismo , Envelhecimento/genética , Western Blotting , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Degeneração Macular/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fagocitose , Epitélio Pigmentado da Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia
18.
Adv Exp Med Biol ; 664: 647-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20238069

RESUMO

Ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF) exhibit multiple biological effects in the developing vertebrate retina. CNTF/LIF inhibits rod photoreceptor, and promotes bipolar cells and Muller glia differentiation. In addition, CNTF/LIF has been shown to have proliferative and apoptotic effects. Moreover, LIF also inhibits retinal vascular development. CNTF/LIF signaling components CNTFRalpha, LIFRbeta, gp130, and a number of STAT proteins are expressed in the retina. CNTF/LIF activates Jak-STAT, ERK, and Notch pathways during retinal development. Perturbation of CNTF induced signal transduction reveals that different combinations of CNTF/LIF signaling pathways regulate differentiation of retinal neurons and glia. Gene expression studies show that CNTF/LIF affects retinogenesis by regulating various genes involved in transcription, signal transduction, protein modification, apoptosis, protein localization, and cell ion homeostasis. Most past studies have deployed ectopic expression or addition of exogenous CNTF/LIF, thus further ana-lysis of mice with conditional mutations in CNTF/LIF signaling components will allow better understanding of in-vivo functions of CNTF/LIF associated signaling events in retinogenesis.


Assuntos
Fator Neurotrófico Ciliar/metabolismo , Fator Inibidor de Leucemia/metabolismo , Organogênese , Retina/embriologia , Retina/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células Fotorreceptoras de Vertebrados , Células Bipolares da Retina , Células-Tronco
19.
Neural Dev ; 4: 32, 2009 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-19686589

RESUMO

BACKGROUND: The paired homeobox protein Pax6 is essential for proliferation and pluripotency of retinal progenitors. However, temporal changes in Pax6 protein expression associated with the generation of various retinal neurons have not been characterized with regard to the cell cycle. Here, we examine the dynamic changes of Pax6 expression among chicken retinal progenitors as they progress through the neurogenic cell cycle, and determine the effects of altered Pax6 levels on retinogenesis. RESULTS: We provide evidence that during the preneurogenic to neurogenic transition, Pax6 protein levels in proliferating progenitor cells are down-regulated. Neurogenic retinal progenitors retain a relatively low level of Pax6 protein, whereas postmitotic neurons either elevate or extinguish Pax6 expression in a cell type-specific manner. Cell imaging and cell cycle analyses show that neurogenic progenitors in the S phase of the cell cycle contain low levels of Pax6 protein, whereas a subset of progenitors exhibits divergent levels of Pax6 protein upon entering the G2 phase of the cell cycle. We also show that M phase cells contain varied levels of Pax6, and some correlate with the onset of early neuronal marker expression, forecasting cell cycle exit and cell fate commitment. Furthermore, either elevating or knocking down Pax6 attenuates cell proliferation and results in increased cell death. Reducing Pax6 decreases retinal ganglion cell genesis and enhances cone photoreceptor and amacrine interneuron production, whereas elevating Pax6 suppresses cone photoreceptor and amacrine cell fates. CONCLUSION: These studies demonstrate for the first time quantitative changes in Pax6 protein expression during the preneurogenic to neurogenic transition and during the neurogenic cell cycle. The results indicate that Pax6 protein levels are stringently controlled in proliferating progenitors. Maintaining a relatively low Pax6 protein level is necessary for S phase re-entry, whereas rapid accumulation or reduction of Pax6 protein during the G2/M phase of the cell cycle may be required for specific neuronal fates. These findings thus provide novel insights on the dynamic regulation of Pax6 protein among neurogenic progenitors and the temporal frame of neuronal fate determination.


Assuntos
Ciclo Celular/fisiologia , Proliferação de Células , Células-Tronco Embrionárias/fisiologia , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Neurogênese/fisiologia , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Retina/citologia , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Morte Celular/genética , Morte Celular/fisiologia , Células Cultivadas , Embrião de Galinha/citologia , Proteínas do Olho/genética , Citometria de Fluxo/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA/fisiologia , Proteínas Repressoras/genética , Retina/embriologia , Transfecção/métodos
20.
Invest Ophthalmol Vis Sci ; 50(10): 4941-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19420345

RESUMO

PURPOSE: Mammalian programmed cell death (PD)-1 is a membrane-associated receptor regulating the balance between T-cell activation, tolerance, and immunopathology; however, its role in neurons has not yet been defined. The hypothesis that PD-1 signaling actively promotes retinal ganglion cell (RGC) death within the developing mouse retina was investigated. METHODS: Mature retinal cell types expressing PD-1 were identified by immunofluorescence staining of vertical retina sections; developmental expression was localized by immunostaining and quantified by Western blot analysis. PD-1 involvement in developmental RGC survival was assessed in vitro using retinal explants and in vivo using PD-1 knockout mice. PD-1 ligand gene expression was detected by RT-PCR. RESULTS: PD-1 is expressed in most adult RGCs and undergoes dynamic upregulation during the early postnatal window of retinal cell maturation and physiological programmed cell death (PCD). In vitro blockade of PD-1 signaling during this time selectively increases the survival of RGCs. Furthermore, PD-1-deficient mice show a selective increase in RGC number in the neonatal retina at the peak of developmental RGC death. Lastly, gene expression of the immune PD-1 ligand genes Pdcd1lg1 and Pdcd1lg2 was found throughout postnatal retina maturation. CONCLUSIONS: These findings collectively support a novel role for a PD-1-mediated signaling pathway in developmental PCD during postnatal RGC maturation.


Assuntos
Antígenos de Superfície/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Apoptose , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/patologia , Animais , Animais Recém-Nascidos , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Western Blotting , Contagem de Células , Sobrevivência Celular , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Ligantes , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/metabolismo , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , RNA Mensageiro/metabolismo , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA