Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 19(11): e1011010, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37930995

RESUMO

Damage to light-sensing photoreceptors (PRs) occurs in highly prevalent retinal diseases. As humans cannot regenerate new PRs, these diseases often lead to irreversible blindness. Intriguingly, animals, such as the zebrafish, can regenerate PRs efficiently and restore functional vision. Upon injury, mature Müller glia (MG) undergo reprogramming to adopt a stem cell-like state. This process is similar to cellular dedifferentiation, and results in the generation of progenitor cells, which, in turn, proliferate and differentiate to replace lost retinal neurons. In this study, we tested whether factors involved in dedifferentiation of Drosophila CNS are implicated in the regenerative response in the zebrafish retina. We found that hairy-related 6 (her6) negatively regulates of PR production by regulating the rate of cell divisions in the MG-derived progenitors. prospero homeobox 1a (prox1a) is expressed in differentiated PRs and may promote PR differentiation through phase separation. Interestingly, upon Her6 downregulation, Prox1a is precociously upregulated in the PRs, to promote PR differentiation; conversely, loss of Prox1a also induces a downregulation of Her6. Together, we identified two novel candidates of PR regeneration that cross regulate each other; these may be exploited to promote human retinal regeneration and vision recovery.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Homeodomínio , Retina , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Regeneração Nervosa/fisiologia , Neuroglia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Homeodomínio/genética
2.
J Neurosci ; 42(26): 5144-5158, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35672150

RESUMO

Photoreceptor degeneration leads to irreversible vision loss in humans with retinal dystrophies such as retinitis pigmentosa. Whereas photoreceptor loss is permanent in mammals, zebrafish possesses the ability to regenerate retinal neurons and restore visual function. Following acute damage, Müller glia (MG) re-enter the cell cycle and produce multipotent progenitors whose progeny differentiate into mature neurons. Both MG reprogramming and proliferation of retinal progenitor cells require reactive microglia and associated inflammatory signaling. Paradoxically, in zebrafish models of retinal degeneration, photoreceptor death does not induce the MG to reprogram and regenerate lost cells. Here, we used male and female zebrafish cep290 mutants to demonstrate that progressive cone degeneration generates an immune response but does not stimulate MG proliferation. Acute light damage triggered photoreceptor regeneration in cep290 mutants but cones were only restored to prelesion densities. Using irf8 mutant zebrafish, we found that the chronic absence of microglia reduced inflammation and rescued cone degeneration in cep290 mutants. Finally, single-cell RNA-sequencing revealed sustained expression of notch3 in MG of cep290 mutants and inhibition of Notch signaling induced MG to re-enter the cell cycle. Our findings provide new insights on the requirements for MG to proliferate and the potential for immunosuppression to prolong photoreceptor survival.SIGNIFICANCE STATEMENT Inherited retinal degenerations (IRDs) are genetic diseases that lead to the progressive loss of photoreceptors and the permanent loss of vision. Zebrafish can regenerate photoreceptors after acute injury by reprogramming Müller glia (MG) into stem-like cells that produce retinal progenitors, but this regenerative process fails to occur in zebrafish models of IRDs. Here, we show that Notch pathway inhibition can promote photoreceptor regeneration in models of progressive degeneration and that immunosuppression can prevent photoreceptor loss. These results offer insight into the pathways that promote MG-dependent regeneration and the role of inflammation in photoreceptor degeneration.


Assuntos
Degeneração Retiniana , Distrofias Retinianas , Animais , Animais Geneticamente Modificados , Proliferação de Células , Feminino , Terapia de Imunossupressão , Inflamação/metabolismo , Masculino , Mamíferos , Regeneração/fisiologia , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Degeneração Retiniana/patologia , Distrofias Retinianas/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
3.
Adv Exp Med Biol ; 1415: 309-317, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440050

RESUMO

Nearly a billion people worldwide are affected by vision-impairing conditions, with retinal degenerative diseases being a major cause of blindness. Unfortunately, such diseases are often permanent and progressive, resulting in further degeneration and loss of sight, due to the human retina possessing little, if any, regenerative capacity. Despite numerous efforts and great progress being made to understand the molecular mechanisms of these diseases and possible therapies, the majority of investigations focused on cell-intrinsic factors. However, the microenvironment surrounding retinal cells throughout these processes also plays an important role, though our current understanding of its involvement remains limited. Here we present a brief overview of the current state of the field of extracellular matrix studies within the retina and its potential roles in retinal diseases and potential therapeutic approaches.


Assuntos
Matriz Extracelular , Degeneração Retiniana , Humanos , Proteínas da Matriz Extracelular , Retina
4.
Exp Eye Res ; 216: 108947, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35074344

RESUMO

Zebrafish possess the ability to completely regenerate the retina following injury, however little is understood about the damage signals that contribute to inducing Müller glia reprogramming and proliferation to regenerate lost neurons. Multiple studies demonstrated that iron contributes to various retinal injuries, however no link has been shown between iron and zebrafish retinal regeneration. Here we demonstrate that Müller glia exhibit transcriptional changes following injury to regulate iron levels within the retina, allowing for increased iron uptake and decreased export. The response of the zebrafish retina to intravitreal iron injection was then characterized, showing that ferrous, and not ferric, iron induces retinal cell death. Additionally, iron chelation resulted in decreased numbers of TUNEL-positive photoreceptors and fewer proliferating Müller glia. Despite the contribution of iron to retinal cell death, inhibition of ferroptosis did not significantly reduce cell death following light treatment. Finally, we demonstrate that both the anti-ferroptotic protein Glutathione peroxidase 4b and the Transferrin receptor 1b are required for Müller glia proliferation following light damage. Together these findings show that iron contributes to cell death in the light-damaged retina and is essential for inducing the Müller glia regeneration response.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Ependimogliais/efeitos dos fármacos , Compostos Ferrosos/toxicidade , Células Fotorreceptoras/efeitos dos fármacos , Lesões Experimentais por Radiação/etiologia , Degeneração Retiniana/induzido quimicamente , Animais , Animais Geneticamente Modificados , Apoptose , Deferiprona/farmacologia , Células Ependimogliais/metabolismo , Marcação In Situ das Extremidades Cortadas , Injeções Intravítreas , Luz , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Células Fotorreceptoras/efeitos da radiação , Lesões Experimentais por Radiação/metabolismo , Receptores da Transferrina/metabolismo , Degeneração Retiniana/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
5.
Glia ; 69(3): 546-566, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32965734

RESUMO

Damage to the zebrafish retina stimulates resident Müller glia to reprogram, reenter the cell cycle, divide asymmetrically, and produce neuronal progenitor cells that amplify and differentiate into the lost neurons. The transition from quiescent to proliferative Müller glia involves both positive and negative regulators. We previously demonstrated that the Notch signaling pathway represses retinal regeneration by maintaining Müller glia quiescence in zebrafish. Here we examine which Notch receptor is necessary to maintain quiescence. Quantitative RT-PCR and RNA-Seq analyses reveal that notch3 is expressed in the undamaged retina and is downregulated in response to light damage. Additionally, Notch3 protein is expressed in quiescent Müller glia of the undamaged retina, is downregulated as Müller glia proliferate, and is reestablished in the Müller glia. Knockdown of Notch3 is sufficient to induce Müller glia proliferation in undamaged retinas and enhances proliferation during light damage. Alternatively, knockdown of Notch1a, Notch1b, or Notch2 decreases the number of proliferating cells during light damage, suggesting that Notch signaling is also required for proliferation during retinal regeneration. We also knockdown the zebrafish Delta and Delta-like proteins, ligands for the Notch receptors, and find that the deltaB morphant possesses an increased number of proliferating cells in the light-damaged retina. As with Notch3, knockdown of DeltaB is sufficient to induce Müller glia proliferation in the absence of light damage. Taken together, the negative regulation of Müller glia proliferation in zebrafish retinal regeneration is mediated by Notch3 and DeltaB.


Assuntos
Retina , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Proliferação de Células , Células Ependimogliais , Neuroglia , Receptor Notch3/genética , Receptores Notch/genética
6.
Glia ; 68(7): 1445-1465, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034934

RESUMO

Brain injury activates complex inflammatory signals in dying neurons, surviving neurons, and glia. Here, we establish that inflammation regulates the regeneration of photoreceptors in the zebrafish retina and determine the cellular expression and function of the inflammatory protease, matrix metalloproteinase 9 (Mmp-9), during this regenerative neurogenesis. Following photoreceptor ablation, anti-inflammatory treatment suppresses the number of injury-induced progenitors and regenerated photoreceptors. Upon photoreceptor injury, mmp-9 is induced in Müller glia and Müller glia-derived photoreceptor progenitors. Deleting mmp-9 results in over production of injury-induced progenitors and regenerated photoreceptors, but over time the absence of Mmp-9 compromises the survival of the regenerated cones. At all time-points studied, the levels of tnf-α are significantly elevated in mutant retinas. Anti-inflammatory treatment in mutants rescues the defects in cone survival. These data provide a link between injury-induced inflammation in the vertebrate CNS, Mmp-9 function during neuronal regeneration and the requirement of Mmp-9 for the survival of regenerated cones.


Assuntos
Inflamação/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Regeneração Nervosa/fisiologia , Regeneração/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Neuroglia/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Células-Tronco/fisiologia , Peixe-Zebra
7.
Exp Eye Res ; 178: 148-159, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30267656

RESUMO

Teleosts are unique in their ability to undergo persistent neurogenesis and to regenerate damaged and lost retinal neurons in adults. This contrasts with the human retina, which is incapable of replacing lost retinal neurons causing vision loss/blindness in the affected individuals. Two cell populations within the adult teleost retina generate new retinal neurons throughout life. Stem cells within the ciliary marginal zone give rise to all retinal cell types except for rod photoreceptors, which are produced by the resident Müller glia that are located within the inner nuclear layer of the entire retina. Understanding the mechanisms that regulate the generation of photoreceptors in the adult teleost retina may ultimately aid developing strategies to overcome vision loss in diseases such as retinitis pigmentosa. Here, we investigated whether photic deprivation alters the proliferative capacity of rod precursor cells, which are generated from Müller glia. In dark-adapted retinas, rod precursor cell proliferation increased, while the number of proliferating Müller glia and their derived olig2:EGFP-positive neuronal progenitor cells was not significantly changed. Cell death of rod photoreceptors was excluded as the inducer of rod precursor cell proliferation, as the number of TUNEL-positive cells and l-plastin-positive microglia in both the outer (ONL) and inner nuclear layer (INL) remained at a similar level throughout the dark-adaptation timecourse. Rod precursor cell proliferation in response to dark-adaptation was characterized by an increased number of EdU-positive cells, i.e. cells that were undergoing DNA replication. These proliferating rod precursor cells in dark-adapted zebrafish differentiated into rod photoreceptors at a comparable percentage and in a similar time frame as those maintained under standard light conditions suggesting that the cell cycle did not stall in dark-adapted retinas. Inhibition of IGF1-receptor signaling reduced the dark-adaptation-mediated proliferation response; however, caloric restriction which has been suggested to be integrated by the IGF1/growth hormone signaling axis did not influence rod precursor cell proliferation in dark-adapted retinas, as similar numbers were observed in starved and normal fed zebrafish. In summary, photic deprivation induces cell cycle entry of rod precursor cells via IGF1-receptor signaling independent of Müller glia proliferation.


Assuntos
Proliferação de Células/efeitos da radiação , Luz , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células-Tronco/citologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/fisiologia , Adaptação à Escuridão , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Marcação In Situ das Extremidades Cortadas , Injeções Intraperitoneais , Neurogênese/fisiologia , Estimulação Luminosa , Pirimidinas/farmacologia , Pirróis/farmacologia , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células-Tronco/metabolismo , Taurina/farmacologia , Peixe-Zebra
8.
Exp Eye Res ; 161: 174-192, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28577895

RESUMO

Sox2 is a well-established neuronal stem cell-associated transcription factor that regulates neural development and adult neurogenesis in vertebrates, and is one of the critical genes used to reprogram differentiated cells into induced pluripotent stem cells. We examined if Sox2 was involved in the early reprogramming-like events that Müller glia undergo as they upregulate many pluripotency- and neural stem cell-associated genes required for proliferation in light-damaged adult zebrafish retinas. In the undamaged adult zebrafish retina, Sox2 is expressed in Müller glia and a subset of amacrine cells, similar to other vertebrates. Following 31 h of light damage, Sox2 expression significantly increased in proliferating Müller glia. Morpholino-mediated knockdown of Sox2 expression resulted in decreased numbers of proliferating Müller glia, while induced overexpression of Sox2 stimulated Müller glia proliferation in the absence of retinal damage. Thus, Sox2 is necessary and sufficient for Müller glia proliferation. We investigated the role of Wnt/ß-catenin signaling, which is a known regulator of sox2 expression during vertebrate retinal development. While ß-catenin 2, but not ß-catenin 1, was necessary for Müller glia proliferation, neither ß-catenin paralog was required for sox2 expression following retinal damage. Sox2 expression was also necessary for ascl1a (neurogenic) and lin28a (reprogramming) expression, but not stat3 expression following retinal damage. Furthermore, Sox2 was required for Müller glial-derived neuronal progenitor cell amplification and expression of the pro-neural marker Tg(atoh7:EGFP). Finally, loss of Sox2 expression prevented complete regeneration of cone photoreceptors. This study is the first to identify a functional role for Sox2 during Müller glial-based regeneration of the vertebrate retina.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células/fisiologia , Células Ependimogliais/metabolismo , Regeneração Nervosa/fisiologia , Proteínas de Ligação a RNA/metabolismo , Retina/fisiologia , Fatores de Transcrição SOX/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Silenciamento de Genes , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Luz , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Lesões Experimentais por Radiação/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Retina/efeitos da radiação , Fatores de Transcrição , Peixe-Zebra
9.
J Neurosci ; 35(47): 15612-34, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26609156

RESUMO

Loss of retinal neurons in adult zebrafish (Danio rerio) induces a robust regenerative response mediated by the reentry of the resident Müller glia into the cell cycle. Upon initiating Müller glia proliferation, their nuclei migrate along the apicobasal axis of the retina in phase with the cell cycle in a process termed interkinetic nuclear migration (INM). We examined the mechanisms governing this cellular process and explored its function in regenerating the adult zebrafish retina. Live-cell imaging revealed that the majority of Müller glia nuclei migrated to the outer nuclear layer (ONL) to divide. These Müller glia formed prominent actin filaments at the rear of nuclei that had migrated to the ONL. Inhibiting actin filament formation or Rho-associated coiled-coil kinase (Rock) activity, which is necessary for phosphorylation of myosin light chain and actin myosin-mediated contraction, disrupted INM with increased numbers of mitotic nuclei remaining in the basal inner nuclear layer, the region where Müller glia typically reside. Double knockdown of Rho-associated coiled-coil kinase 2a (Rock2a) and Rho-associated coiled-coil kinase 2b (Rock2b) similarly disrupted INM and reduced Müller glial cell cycle reentry. In contrast, Rock inhibition immediately before the onset of INM did not affect Müller glia proliferation, but subsequently reduced neuronal progenitor cell proliferation due to early cell cycle exit. Long-term, Rock inhibition increased the generation of mislocalized ganglion/amacrine cells at the expense of rod and cone photoreceptors. In summary, INM is driven by an actin-myosin-mediated process controlled by Rock2a and Rock2b activity, which is required for sufficient proliferation and regeneration of photoreceptors after light damage. SIGNIFICANCE STATEMENT: The human retina does not replace lost or damaged neurons, ultimately causing vision impairment. In contrast, zebrafish are capable of regenerating lost neurons. Understanding the mechanisms that regulate retinal regeneration in these organisms will help to elucidate approaches to stimulate a similar response in humans. In the damaged zebrafish retina, Müller glia dedifferentiate and proliferate to generate neuronal progenitor cells (NPCs) that differentiate into the lost neurons. We show that the nuclei of Müller glia and NPCs migrate apically and basally in phase with the cell cycle. This migration is facilitated by the actin cytoskeleton and Rho-associated coiled-coil kinases (Rocks). We demonstrate that Rock function is required for sufficient proliferation and the regeneration of photoreceptors, likely via regulating nuclear migration.


Assuntos
Actinas/fisiologia , Movimento Celular/fisiologia , Núcleo Celular/fisiologia , Citoesqueleto/fisiologia , Regeneração Nervosa/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Quinases Associadas a rho/fisiologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Células Cultivadas , Feminino , Masculino , Retina/citologia , Retina/fisiologia , Peixe-Zebra
10.
Trends Genet ; 29(11): 611-20, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23927865

RESUMO

For centuries, philosophers and scientists have been fascinated by the principles and implications of regeneration in lower vertebrate species. Two features have made zebrafish an informative model system for determining mechanisms of regenerative events. First, they are highly regenerative, able to regrow amputated fins, as well as a lesioned brain, retina, spinal cord, heart, and other tissues. Second, they are amenable to both forward and reverse genetic approaches, with a research toolset regularly updated by an expanding community of zebrafish researchers. Zebrafish studies have helped identify new mechanistic underpinnings of regeneration in multiple tissues and, in some cases, have served as a guide for contemplating regenerative strategies in mammals. Here, we review the recent history of zebrafish as a genetic model system for understanding how and why tissue regeneration occurs.


Assuntos
Nadadeiras de Animais/fisiologia , Encéfalo/fisiologia , Coração/fisiologia , Regeneração , Retina/fisiologia , Medula Espinal/fisiologia , Animais , Modelos Animais , Peixe-Zebra/genética
11.
Adv Exp Med Biol ; 854: 587-93, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427463

RESUMO

In the adult zebrafish, death of retinal neurons stimulates Müller glia to re-enter the cell cycle to produce neuronal progenitor cells (NPCs) that undergo further cell divisions and differentiate to replace lost neurons in the correct spatial locations. Understanding the mechanisms regulating retinal regeneration will ultimately provide avenues to overcome vision loss in human. Recently, the observation of interkinetic nuclear migration (INM) of Müller glia in the regenerating zebrafish retina resulted in the inclusion of an additional complex step to the regeneration process. The pathways regulating INM and its function in the regenerating retina have not been well studied. Here, we summarize the evidence for INM in the regenerating retina and review mechanisms that control INM during neuro-epithelial development in the context of pathways known to be critical during retinal regeneration.


Assuntos
Movimento Celular/fisiologia , Células Ependimogliais/fisiologia , Retina/fisiologia , Neurônios Retinianos/fisiologia , Animais , Núcleo Celular/fisiologia , Humanos , Neurogênese , Regeneração , Retina/citologia , Peixe-Zebra/fisiologia
12.
J Neurosci ; 34(43): 14403-19, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339752

RESUMO

Retinal damage in teleosts, unlike mammals, induces robust Müller glia-mediated regeneration of lost neurons. We examined whether Notch signaling regulates Müller glia proliferation in the adult zebrafish retina and demonstrated that Notch signaling maintains Müller glia in a quiescent state in the undamaged retina. Repressing Notch signaling, through injection of the γ-secretase inhibitor RO4929097, stimulates a subset of Müller glia to reenter the cell cycle without retinal damage. This RO4929097-induced Müller glia proliferation is mediated by repressing Notch signaling because inducible expression of the Notch Intracellular Domain (NICD) can reverse the effect. This RO4929097-induced proliferation requires Ascl1a expression and Jak1-mediated Stat3 phosphorylation/activation, analogous to the light-damaged retina. Moreover, coinjecting RO4929097 and TNFα, a previously identified damage signal, induced the majority of Müller glia to reenter the cell cycle and produced proliferating neuronal progenitor cells that committed to a neuronal lineage in the undamaged retina. This demonstrates that repressing Notch signaling and activating TNFα signaling are sufficient to induce Müller glia proliferation that generates neuronal progenitor cells that differentiate into retinal neurons, mimicking the responses observed in the regenerating retina.


Assuntos
Proliferação de Células/fisiologia , Células Ependimogliais/fisiologia , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/fisiologia , Receptores Notch/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Animais Geneticamente Modificados , Proliferação de Células/efeitos dos fármacos , Células Ependimogliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Masculino , Regeneração Nervosa/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Peixe-Zebra
13.
Dev Biol ; 392(2): 393-403, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24858486

RESUMO

Damage of the zebrafish retina triggers a spontaneous regeneration response that is initiated by Müller Glia (MG) dedifferentiation and asymmetric cell division to produce multipotent progenitor cells. Subsequent expansion of the progenitor pool by proliferation is critical for retina regeneration. Pax6b expression in the progenitor cells is necessary for their proliferation, but exact regulation of its expression is unclear. Here, we show that miR-203 is downregulated during regeneration in proliferating progenitor cells. Elevated miR-203 levels inhibit progenitor cell expansion without affecting MG dedifferentiation or progenitor cell generation. Using GFP-reporter assays and gain and loss of function experiments in the retina, we show that miR-203 expression must be suppressed to allow pax6b expression and subsequent progenitor cell proliferation.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica/genética , MicroRNAs/metabolismo , Regeneração/fisiologia , Retina/fisiologia , Células-Tronco/fisiologia , Peixe-Zebra/fisiologia , Animais , Western Blotting , Clonagem Molecular , Eletroporação , Citometria de Fluxo , Imuno-Histoquímica , MicroRNAs/genética , Microinjeções , Morfolinos/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Peixe-Zebra/genética
14.
J Biol Chem ; 289(10): 6934-6940, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24469449

RESUMO

The human oncogene SCL/TAL1 interrupting locus (Stil) is highly conserved in vertebrate species. Previously, we identified a homolog of the Stil gene in zebrafish mutant (night blindness b, nbb), which showed neural defects in the retina (e.g. dopaminergic cell degeneration and/or lack of regeneration). In this research, we examined the roles of Stil in cell proliferation after degeneration in adult zebrafish retinas. We demonstrated that knockdown of Stil gene expression or inhibition of Sonic hedgehog (Shh) signaling transduction decreases the rate of cell proliferation. In contrast, activation of Shh signal transduction promotes cell proliferation. In nbb(+/-) retinas, inhibition of SUFU (a repressor in the Shh pathway) rescues the defects in cell proliferation due to down-regulation of Stil gene expression. The latter data suggest that Stil play a role in cell proliferation through the Shh signal transduction pathway.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proliferação de Células , Proteínas Hedgehog/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Retina/patologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Células Ependimogliais/patologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/genética , Oxidopamina/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Retina/efeitos dos fármacos , Retina/metabolismo , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/patologia , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Transdução de Sinais , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
Dev Dyn ; 243(12): 1591-605, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25220904

RESUMO

BACKGROUND: Adult zebrafish spontaneously regenerate their retinas after damage. Although a number of genes and signaling pathways involved in regeneration have been identified, the exact mechanisms regulating various aspects of regeneration are unclear. microRNAs (miRNAs) were examined for their potential roles in regulating zebrafish retinal regeneration. RESULTS: To investigate the requirement of miRNAs during zebrafish retinal regeneration, we knocked down the expression of Dicer in retinas prior to light-induced damage. Reduced Dicer expression significantly decreased the number of proliferating Müller glia-derived neuronal progenitor cells during regeneration. To identify individual miRNAs with roles in neuronal progenitor cell proliferation, we collected retinas at different stages of light damage and performed small RNA high-throughput sequencing. We identified subsets of miRNAs that were differentially expressed during active regeneration but returned to basal levels once regeneration was completed. We then knocked down five different miRNAs that increased in expression and assessed the effects on retinal regeneration. Reduction of miR-142b and miR-146a expression significantly reduced INL proliferation at 51 h of light treatment, while knockdown of miR-7a, miR-27c, and miR-31 expression significantly reduced INL proliferation at 72 h of constant light. CONCLUSIONS: miRNAs exhibit dynamic expression profiles during retinal regeneration and are necessary for neuronal progenitor cell proliferation.


Assuntos
Proliferação de Células/fisiologia , Regulação da Expressão Gênica , MicroRNAs/biossíntese , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Regeneração/fisiologia , Retina/fisiologia , Ribonuclease III/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/metabolismo , Animais , Técnicas de Silenciamento de Genes , MicroRNAs/genética , Ribonuclease III/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
J Neurosci ; 33(15): 6524-39, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575850

RESUMO

Intense light exposure causes photoreceptor apoptosis in dark-adapted adult albino zebrafish (Danio rerio). Subsequently, Müller glia increase expression of the Achaete-scute complex-like 1a (Ascl1a) and Signal transducer and activator of transcription 3 (Stat3) transcription factors and re-enter the cell cycle to yield undifferentiated neuronal progenitors that continue to proliferate, migrate to the outer nuclear layer, and differentiate into photoreceptors. A proteomic analysis of light-damaged retinal homogenates, which induced Müller glia proliferation when injected into an undamaged eye, revealed increased expression of tumor necrosis factor α (TNFα) signaling proteins relative to undamaged retinal homogenates. TNFα expression initially increased in apoptotic photoreceptors and later in Müller glia. Morpholino-mediated knockdown of TNFα expression before light damage diminished the expression of both Ascl1a and Stat3 in Müller glia and significantly reduced the number of proliferating Müller glia without affecting photoreceptor cell death. Knockdown of TNFα expression in the Müller glia resulted in fewer proliferating Müller glia, suggesting that Müller glial-derived TNFα recruited additional Müller glia to re-enter the cell cycle. While TNFα is required for increased Ascl1a and Stat3 expression, Ascl1a and Stat3 are both necessary for TNFα expression in Müller glia. Apoptotic inner retinal neurons, resulting from intravitreal injection of ouabain, also exhibited increased TNFα expression that was required for Müller glia proliferation. Thus, TNFα is the first molecule identified that is produced by dying retinal neurons and is necessary to induce Müller glia to proliferate in the zebrafish retinal regeneration response.


Assuntos
Apoptose/fisiologia , Proliferação de Células/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Neuroglia/fisiologia , Neurônios Retinianos/metabolismo , Neurônios Retinianos/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/fisiologia , Animais , Apoptose/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes/métodos , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Luz/efeitos adversos , Regeneração Nervosa/efeitos dos fármacos , Ouabaína/farmacologia , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Neurônios Retinianos/efeitos dos fármacos , Fator de Transcrição STAT3/biossíntese , Fatores de Transcrição , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
17.
Exp Eye Res ; 123: 131-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23880528

RESUMO

This article examines our current knowledge underlying the mechanisms involved in neuronal regeneration in the adult zebrafish retina. Zebrafish, which has the capacity to regenerate a wide variety of tissues and organs (including the fins, kidney, heart, brain, and spinal cord), has become the premier model system to study retinal regeneration due to the robustness and speed of the response and the variety of genetic tools that can be applied to study this question. It is now well documented that retinal damage induces the resident Müller glia to dedifferentiate and reenter the cell cycle to produce neuronal progenitor cells that continue to proliferate, migrate to the damaged retinal layer and differentiate into the missing neuronal cell types. Increasing our understanding of how these cellular events are regulated and occur in response to neuronal damage may provide critical information that can be applied to stimulating a regeneration response in the mammalian retina. In this review, we will focus on the genes/proteins that regulate zebrafish retinal regeneration and will attempt to critically evaluate how these factors may interact to correctly orchestrate the definitive cellular events that occur during regeneration.


Assuntos
Células Ependimogliais/metabolismo , Regeneração/fisiologia , Doenças Retinianas/fisiopatologia , Neurônios Retinianos/fisiologia , Peixe-Zebra/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células-Tronco Neurais/citologia
18.
Adv Exp Med Biol ; 801: 535-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24664741

RESUMO

In a screen to identify zebrafish eye mutants, we isolated the good effort (gef) mutant. The retina of gef embryos is characterized by the successful initiation of the optic primordium and normal retinal development over the first 2 days post fertilization (dpf). The mutant retina, however, fails to continue to grow. Embryos from gef heterozygous incrosses were analyzed for cell death by acridine orange and by TUNEL labeling at 2 dpf. Significantly more TUNEL-positive and acridine orange-labeled dying cells were found in gef mutant embryos at 2 dpf relative to wild-type embryos. Because this time was earlier than any observable gross morphological differences, this cell death was likely the cause of the gross morphological defects. Meiotic mapping localized the mutation interval to a one-megabase interval on zebrafish chromosome 9.


Assuntos
Mapeamento Cromossômico/métodos , Degeneração Retiniana/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Morte Celular/fisiologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose/fisiologia , Fenótipo , Retina/embriologia , Retina/fisiologia
19.
Front Cell Dev Biol ; 12: 1406330, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38938553

RESUMO

Zebrafish possess the ability to regenerate dying neurons in response to retinal injury, with both Müller glia and microglia playing integral roles in this response. Resident Müller glia respond to damage by reprogramming and undergoing an asymmetric cell division to generate a neuronal progenitor cell, which continues to proliferate and differentiate into the lost neurons. In contrast, microglia become reactive, phagocytose dying cells, and release inflammatory signals into the surrounding tissue following damage. In recent years, there has been increased attention on elucidating the role that microglia play in regulating retinal regeneration. Here we demonstrate that inflammatory cytokines are differentially expressed during retinal regeneration, with the expression of a subset of pro-inflammatory cytokine genes upregulated shortly after light damage and the expression of a different subset of cytokine genes subsequently increasing. We demonstrate that both cytokine IL-1ß and IL-10 are essential for Müller glia proliferation in the light-damaged retina. While IL-1ß is sufficient to induce Müller glia proliferation in an undamaged retina, expression of IL-10 in undamaged retinas only induces Müller glia to express gliotic markers. Together, these findings demonstrate the essential role of inflammatory cytokines IL-1ß and IL-10 on Müller glia proliferation following light damage in adult zebrafish.

20.
Nature ; 446(7137): 797-800, 2007 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-17392791

RESUMO

The development of cell polarity is an essential prerequisite for tissue morphogenesis during embryogenesis, particularly in the development of epithelia. In addition, oriented cell division can have a powerful influence on tissue morphogenesis. Here we identify a novel mode of polarized cell division that generates pairs of neural progenitors with mirror-symmetric polarity in the developing zebrafish neural tube and has dramatic consequences for the organization of embryonic tissue. We show that during neural rod formation the polarity protein Pard3 is localized to the cleavage furrow of dividing progenitors, and then mirror-symmetrically inherited by the two daughter cells. This allows the daughter cells to integrate into opposite sides of the developing neural tube. Furthermore, these mirror-symmetric divisions have powerful morphogenetic influence: when forced to occur in ectopic locations during neurulation, they orchestrate the development of mirror-image pattern formation and the consequent generation of ectopic neural tubes.


Assuntos
Padronização Corporal , Polaridade Celular , Células Epiteliais/citologia , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Neurônios/citologia , Peixe-Zebra/embriologia , Animais , Proteínas de Transporte/metabolismo , Divisão Celular , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA