Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Methods Mol Biol ; 2636: 367-388, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36881311

RESUMO

Following retinal injury, zebrafish possess the remarkable capacity to endogenously regenerate lost retinal neurons from Müller glia-derived neuronal progenitor cells. Additionally, neuronal cell types that are undamaged and persist in the injured retina are also produced. Thus, the zebrafish retina is an excellent system to study the integration of all neuronal cell types into an existing neuronal circuit. The few studies that examined axonal/dendritic outgrowth and the establishment of synaptic contacts by regenerated neurons predominantly utilized fixed tissue samples. We recently established a flatmount culture model to monitor Müller glia nuclear migration in real time by two-photon microscopy. However, in retinal flatmounts, z-stacks of the entire retinal z-dimension have to be acquired to image cells that extend through parts or the entirety of the neural retina, such as bipolar cells and Müller glia, respectively. Cellular processes with fast kinetics might thus be missed. Therefore, we generated a retinal cross-section culture from light-damaged zebrafish to image the entire Müller glia in one z-plane. Isolated dorsal retinal hemispheres were cut into two dorsal quarters and mounted with the cross-section view facing the coverslips of culture dishes, which allowed monitoring Müller glia nuclear migration using confocal microscopy. Confocal imaging of cross-section cultures is ultimately also applicable to live cell imaging of axon/dendrite formation of regenerated bipolar cells, while the flatmount culture model will be more suitable to monitor axon outgrowth of ganglion cells.


Assuntos
Neurônios Retinianos , Peixe-Zebra , Animais , Retina , Neuroglia , Microscopia Confocal
7.
Antioxidants (Basel) ; 12(9)2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37759997

RESUMO

Choroideremia (CHM) is a rare X-linked chorioretinal dystrophy, affecting the photoreceptors, retinal pigment epithelium (RPE) and choroid, with no approved therapy. CHM is caused by mutations in the CHM gene, which encodes the ubiquitously expressed Rab escort protein 1 (REP1). REP1 is involved in prenylation, a post-translational modification of Rab proteins, and plays an essential role in intracellular trafficking. In this study, we examined oxidative and endoplasmic reticulum (ER) stress pathways in chmru848 zebrafish and CHMY42X patient fibroblasts, and screened a number of neuroprotectants for their ability to reduce stress. The expression of the oxidative stress markers txn, cat and sod3a, and the ER stress markers bip, atf4 and atf6, were dysregulated in chmru848 fish. The expression of SOD2 was also reduced in CHMY42X fibroblasts, along with reduced BIP and increased CHOP expression. The lack of REP1 is associated with defects in vesicular trafficking, photoreceptor outer segment phagocytosis and melanosome transport, leading to increased levels of stress within the retina and RPE. Drugs targeting oxidative and ER stress pathways represent novel therapeutic avenues.

8.
Life Sci Alliance ; 6(11)2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37643867

RESUMO

Enhancers play a critical role in development by precisely modulating spatial, temporal, and cell type-specific gene expression. Sequence variants in enhancers have been implicated in diseases; however, establishing the functional consequences of these variants is challenging because of a lack of understanding of precise cell types and developmental stages where the enhancers are normally active. PAX6 is the master regulator of eye development, with a regulatory landscape containing multiple enhancers driving the expression in the eye. Whether these enhancers perform additive, redundant or distinct functions is unknown. Here, we describe the precise cell types and regulatory activity of two PAX6 retinal enhancers, HS5 and NRE. Using a unique combination of live imaging and single-cell RNA sequencing in dual enhancer-reporter zebrafish embryos, we uncover differences in the spatiotemporal activity of these enhancers. Our results show that although overlapping, these enhancers have distinct activities in different cell types and therefore likely nonredundant functions. This work demonstrates that unique cell type-specific activities can be uncovered for apparently similar enhancers when investigated at high resolution in vivo.


Assuntos
Fator de Transcrição PAX6 , Sequências Reguladoras de Ácido Nucleico , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Retina , Fator de Transcrição PAX6/genética , Proteínas de Peixe-Zebra/genética
9.
Indian J Ophthalmol ; 70(12): 4119-4129, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36453299

RESUMO

Aniridia is a pan-ocular genetic developmental eye disorder characterized by complete or partial iris and foveal hypoplasia, for which there is no treatment currently. Progressive sight loss can arise from cataracts, glaucoma, and aniridia-related keratopathy, which can be managed conservatively or through surgical intervention. The vast majority of patients harbor heterozygous mutations involving the PAX6 gene, which is considered the master transcription factor of early eye development. Over the past decades, several disease models have been investigated to gain a better understanding of the molecular pathophysiology, including several mouse and zebrafish strains and, more recently, human-induced pluripotent stem cells (hiPSCs) derived from aniridia patients. The latter provides a more faithful cellular system to study early human eye development. This review outlines the main aniridia-related animal and cellular models used to study aniridia and highlights the key discoveries that are bringing us closer to a therapy for patients.


Assuntos
Aniridia , Catarata , Glaucoma , Humanos , Animais , Camundongos , Peixe-Zebra , Aniridia/diagnóstico , Aniridia/genética , Iris , Fator de Transcrição PAX6/genética
10.
Biomedicines ; 9(8)2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34440066

RESUMO

Blunt-force traumatic brain injury (TBI) affects an increasing number of people worldwide as the range of injury severity and heterogeneity of injury pathologies have been recognized. Most current damage models utilize non-regenerative organisms, less common TBI mechanisms (penetrating, chemical, blast), and are limited in scalability of injury severity. We describe a scalable blunt-force TBI model that exhibits a wide range of human clinical pathologies and allows for the study of both injury pathology/progression and mechanisms of regenerative recovery. We modified the Marmarou weight drop model for adult zebrafish, which delivers a scalable injury spanning mild, moderate, and severe phenotypes. Following injury, zebrafish display a wide range of severity-dependent, injury-induced pathologies, including seizures, blood-brain barrier disruption, neuroinflammation, edema, vascular injury, decreased recovery rate, neuronal cell death, sensorimotor difficulties, and cognitive deficits. Injury-induced pathologies rapidly dissipate 4-7 days post-injury as robust cell proliferation is observed across the neuroaxis. In the cerebellum, proliferating nestin:GFP-positive cells originated from the cerebellar crest by 60 h post-injury, which then infiltrated into the granule cell layer and differentiated into neurons. Shh pathway genes increased in expression shortly following injury. Injection of the Shh agonist purmorphamine in undamaged fish induced a significant proliferative response, while the proliferative response was inhibited in injured fish treated with cyclopamine, a Shh antagonist. Collectively, these data demonstrate that a scalable blunt-force TBI to adult zebrafish results in many pathologies similar to human TBI, followed by recovery, and neuronal regeneration in a Shh-dependent manner.

11.
Purinergic Signal ; 6(2): 189-200, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20806011

RESUMO

UNLABELLED: Intercellular Ca(2+) waves can coordinate the action of large numbers of cells over significant distances. Recent work in many different systems has indicated that the release of ATP is fundamental for the propagation of most Ca(2+) waves. In the organ of hearing, the cochlea, ATP release is involved in critical signalling events during tissue maturation. ATP-dependent signalling is also implicated in the normal hearing process and in sensing cochlear damage. Here, we show that two distinct Ca(2+) waves are triggered during damage to cochlear explants. Both Ca(2+) waves are elicited by extracellular ATP acting on P2 receptors, but they differ in their source of Ca(2+), their velocity, their extent of spread and the cell type through which they propagate. A slower Ca(2+) wave (14 mum/s) communicates between Deiters' cells and is mediated by P2Y receptors and Ca(2+) release from IP(3)-sensitive stores. In contrast, a faster Ca(2+) wave (41 mum/s) propagates through sensory hair cells and is mediated by Ca(2+) influx from the external environment. Using inhibitors and selective agonists of P2 receptors, we suggest that the faster Ca(2+) wave is mediated by P2X(4) receptors. Thus, in complex tissues, the expression of different receptors determines the propagation of distinct intercellular communication signals. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11302-010-9193-8) contains supplementary material, which is available to authorized users.

12.
Front Cell Dev Biol ; 8: 617923, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33598455

RESUMO

Adult zebrafish possess the remarkable capacity to regenerate neurons. In the damaged zebrafish retina, Müller glia reprogram and divide to produce neuronal progenitor cells (NPCs) that proliferate and differentiate into both lost neuronal cell types and those unaffected by the damage stimulus, which suggests that developmental specification/differentiation programs might be recapitulated during regeneration. Quantitative real-time polymerase chain reaction revealed that developmental competence factors are expressed following photoreceptor damage induced by intense light or in a genetic rod photoreceptor cell ablation model. In both light- and N-Methyl-D-aspartic acid (NMDA)-damaged adult zebrafish retinas, NPCs, but not proliferating Müller glia, expressed fluorescent reporters controlled by promoters of ganglion (atoh7), amacrine (ptf1a), bipolar (vsx1), or red cone photoreceptor cell competence factors (thrb) in a temporal expression sequence. In both damage paradigms, atoh7:GFP was expressed first, followed by ptf1a:EGFP and lastly, vsx1:GFP, whereas thrb:Tomato was observed in NPCs at the same time as ptf1a:GFP following light damage but shifted alongside vsx1:GFP in the NMDA-damaged retina. Moreover, HuC/D, indicative of ganglion and amacrine cell differentiation, colocalized with atoh7:GFP prior to ptf1a:GFP expression in the ganglion cell layer, which was followed by Zpr-1 expression (red/green cone photoreceptors) in thrb:Tomato-positive cells in the outer nuclear layer in both damage paradigms, mimicking the developmental differentiation sequence. However, comparing NMDA- to light-damaged retinas, the fraction of PCNA-positive cells expressing atoh7:GFP increased, that of thrb:Tomato and vsx1:GFP decreased, and that of ptf1a:GFP remained similar. To summarize, developmental cell specification programs were recapitulated during retinal regeneration, which adapted to account for the cell type lost.

13.
Annu Rev Vis Sci ; 6: 171-193, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32343929

RESUMO

In humans, various genetic defects or age-related diseases, such as diabetic retinopathies, glaucoma, and macular degeneration, cause the death of retinal neurons and profound vision loss. One approach to treating these diseases is to utilize stem and progenitor cells to replace neurons in situ, with the expectation that new neurons will create new synaptic circuits or integrate into existing ones. Reprogramming non-neuronal cells in vivo into stem or progenitor cells is one strategy for replacing lost neurons. Zebrafish have become a valuable model for investigating cellular reprogramming and retinal regeneration. This review summarizes our current knowledge regarding spontaneous reprogramming of Müller glia in zebrafish and compares this knowledge to research efforts directed toward reprogramming Müller glia in mammals. Intensive research using these animal models has revealed shared molecular mechanisms that make Müller glia attractive targets for cellular reprogramming and highlighted the potential for curing degenerative retinal diseases from intrinsic cellular sources.


Assuntos
Células Ependimogliais/fisiologia , Regeneração Nervosa/fisiologia , Neurônios Retinianos/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Metilação de DNA , Epigenômica , Humanos , Receptores Notch/metabolismo , Neurônios Retinianos/citologia , Transdução de Sinais , Células-Tronco , Peixe-Zebra
14.
Science ; 370(6519)2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33004674

RESUMO

Injury induces retinal Müller glia of certain cold-blooded vertebrates, but not those of mammals, to regenerate neurons. To identify gene regulatory networks that reprogram Müller glia into progenitor cells, we profiled changes in gene expression and chromatin accessibility in Müller glia from zebrafish, chick, and mice in response to different stimuli. We identified evolutionarily conserved and species-specific gene networks controlling glial quiescence, reactivity, and neurogenesis. In zebrafish and chick, the transition from quiescence to reactivity is essential for retinal regeneration, whereas in mice, a dedicated network suppresses neurogenic competence and restores quiescence. Disruption of nuclear factor I transcription factors, which maintain and restore quiescence, induces Müller glia to proliferate and generate neurons in adult mice after injury. These findings may aid in designing therapies to restore retinal neurons lost to degenerative diseases.


Assuntos
Reprogramação Celular/genética , Células Ependimogliais/citologia , Redes Reguladoras de Genes , Regeneração Nervosa/genética , Neurogênese/genética , Animais , Galinhas , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , RNA-Seq , Peixe-Zebra
15.
J Neurosci ; 28(19): 4918-28, 2008 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-18463245

RESUMO

Acoustic overstimulation and ototoxic drugs can cause permanent hearing loss as a result of the damage and death of cochlear hair cells. Relatively little is known about the signaling pathways triggered by such trauma, although a significant role has been described for the c-Jun N-terminal kinase [one of the mitogen-activated protein kinases (MAPKs)] pathway. We investigated the role of another MAPK family, the extracellularly regulated kinases 1 and 2 (ERK1/2) during hair cell damage in neonatal cochlear explants. Within minutes of subjecting explants to mechanical damage, ERK1/2 were transiently activated in Deiters' and phalangeal cells but not in hair cells. The activation of ERK1/2 spread along the length of the cochlea, reaching its peak 5-10 min after damage onset. Release of extracellular ATP and the presence of functional connexin proteins were critical for the activation and spread of ERK1/2. Damage elicited an intercellular Ca(2+) wave in the hair cell region in the first seconds after damage. In the absence of Ca(2+) influx, the intercellular Ca(2+) wave and the magnitude and spread of ERK1/2 activation were reduced. Treatment with the aminoglycoside neomycin produced a similar pattern of ERK1/2 activation in supporting cells surrounding pyknotic hair cells. When ERK1/2 activation was prevented, there was a reduction in the number of pyknotic hair cells. Thus, activation of ERK1/2 in cochlear supporting cells in vitro is a common damage signaling mechanism that acts to promote hair cell death, indicating a direct role for supporting cells in regulating hair cell death.


Assuntos
Cóclea/lesões , Cóclea/fisiopatologia , Células Ciliadas Auditivas , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Morte Celular , Cóclea/efeitos dos fármacos , Cóclea/enzimologia , Conexinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Líquido Extracelular/metabolismo , Células Ciliadas Auditivas/efeitos dos fármacos , Técnicas In Vitro , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neomicina/farmacologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Distribuição Tecidual
16.
Elife ; 72018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30475208

RESUMO

Nephron segmentation involves a concert of genetic and molecular signals that are not fully understood. Through a chemical screen, we discovered that alteration of peroxisome proliferator-activated receptor (PPAR) signaling disrupts nephron segmentation in the zebrafish embryonic kidney (Poureetezadi et al., 2016). Here, we show that the PPAR co-activator ppargc1a directs renal progenitor fate. ppargc1a mutants form a small distal late (DL) segment and an expanded proximal straight tubule (PST) segment. ppargc1a promotes DL fate by regulating the transcription factor tbx2b, and restricts expression of the transcription factor sim1a to inhibit PST fate. Interestingly, sim1a restricts ppargc1a expression to promote the PST, and PST development is fully restored in ppargc1a/sim1a-deficient embryos, suggesting Ppargc1a and Sim1a counterbalance each other in an antagonistic fashion to delineate the PST segment boundary during nephrogenesis. Taken together, our data reveal new roles for Ppargc1a during development, which have implications for understanding renal birth defects.


Assuntos
Padronização Corporal , Néfrons/embriologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Bezafibrato/farmacologia , Padronização Corporal/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Testes Genéticos , Morfolinos/farmacologia , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/química , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Fenótipo , Pronefro/efeitos dos fármacos , Pronefro/embriologia , Pronefro/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
17.
Neural Regen Res ; 12(8): 1210-1219, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28966629

RESUMO

Sensing and responding to our environment requires functional neurons that act in concert. Neuronal cell loss resulting from degenerative diseases cannot be replaced in humans, causing a functional impairment to integrate and/or respond to sensory cues. In contrast, zebrafish (Danio rerio) possess an endogenous capacity to regenerate lost neurons. Here, we will focus on the processes that lead to neuronal regeneration in the zebrafish retina. Dying retinal neurons release a damage signal, tumor necrosis factor α, which induces the resident radial glia, the Müller glia, to reprogram and re-enter the cell cycle. The Müller glia divide asymmetrically to produce a Müller glia that exits the cell cycle and a neuronal progenitor cell. The arising neuronal progenitor cells undergo several rounds of cell divisions before they migrate to the site of damage to differentiate into the neuronal cell types that were lost. Molecular and immunohistochemical studies have predominantly provided insight into the mechanisms that regulate retinal regeneration. However, many processes during retinal regeneration are dynamic and require live-cell imaging to fully discern the underlying mechanisms. Recently, a multiphoton imaging approach of adult zebrafish retinal cultures was developed. We will discuss the use of live-cell imaging, the currently available tools and those that need to be developed to advance our knowledge on major open questions in the field of retinal regeneration.

18.
J Vis Exp ; (120)2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28287581

RESUMO

An endogenous regeneration program is initiated by Müller glia in the adult zebrafish (Danio rerio) retina following neuronal damage and death. The Müller glia re-enter the cell cycle and produce neuronal progenitor cells that undergo subsequent rounds of cell divisions and differentiate into the lost neuronal cell types. Both Müller glia and neuronal progenitor cell nuclei replicate their DNA and undergo mitosis in distinct locations of the retina, i.e. they migrate between the basal Inner Nuclear Layer (INL) and the Outer Nuclear Layer (ONL), respectively, in a process described as Interkinetic Nuclear Migration (INM). INM has predominantly been studied in the developing retina. To examine the dynamics of INM in the adult regenerating zebrafish retina in detail, live-cell imaging of fluorescently-labeled Müller glia/neuronal progenitor cells is required. Here, we provide the conditions to isolate and culture dorsal retinas from Tg[gfap:nGFP]mi2004 zebrafish that were exposed to constant intense light for 35 h. We also show that these retinal cultures are viable to perform live-cell imaging experiments, continuously acquiring z-stack images throughout the thickness of the retinal explant for up to 8 h using multiphoton microscopy to monitor the migratory behavior of gfap:nGFP-positive cells. In addition, we describe the details to perform post-imaging analysis to determine the velocity of apical and basal INM. To summarize, we established conditions to study the dynamics of INM in an adult model of neuronal regeneration. This will advance our understanding of this crucial cellular process and allow us to determine the mechanisms that control INM.


Assuntos
Microscopia de Fluorescência por Excitação Multifotônica/métodos , Retina/citologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Microscopia , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA