Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Adv Exp Med Biol ; 1074: 327-333, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721960

RESUMO

Humans with mutations in the phototransduction pathway develop forms of retinal degeneration, such as retinitis pigmentosa, cone dystrophy, or Leber congenital amaurosis. Similarly, numerous phototransduction mutant animal models resemble retinal degeneration. In our lab, using a zebrafish model, we study cone-specific phototransduction mutants. cGMP is the second messenger in the phototransduction pathway, and abnormal cGMP levels are associated with photoreceptor death. Rd1, a rod-specific phosphodiesterase 6 (Pde6) subunit mutant in mice, is one of the most widely used animal models for retinal degeneration. Rd1 mutant mice accumulate cGMP, causing rapid photoreceptor degeneration. However, much less is known about photoreceptor mutants producing abnormally low levels of cGMP. Here, focusing on Pde6 mutants in zebrafish and mice, we propose a correlation between cGMP levels and speed of photoreceptor degeneration.


Assuntos
GMP Cíclico/fisiologia , Modelos Animais de Doenças , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/metabolismo , Animais , Defeitos da Visão Cromática/enzimologia , Defeitos da Visão Cromática/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/deficiência , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/fisiologia , Proteínas do Olho , Previsões , Humanos , Transdução de Sinal Luminoso , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneração Retiniana/genética , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
2.
J Neurogenet ; 31(3): 88-101, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28812418

RESUMO

Zebrafish are an excellent animal model for research on vertebrate development and human diseases. Sophisticated genetic tools including large-scale mutagenesis methodology make zebrafish useful for studying neuronal degenerative diseases. Here, we review zebrafish models of inherited ophthalmic diseases, focusing on cGMP metabolism in photoreceptors. cGMP is the second messenger of phototransduction, and abnormal cGMP levels are associated with photoreceptor death. cGMP concentration represents a balance between cGMP phosphodiesterase 6 (PDE6) and guanylate cyclase (GC) activities in photoreceptors. Various zebrafish cGMP metabolism mutants were used to clarify molecular mechanisms by which dysfunctions in this pathway trigger photoreceptor degeneration. Here, we review the history of research on the retinal degeneration (rd) mutant mouse, which carries a genetic mutation of PDE6b, and we also highlight recent research in photoreceptor degeneration using zebrafish models. Several recent discoveries that provide insight into cGMP toxicity in photoreceptors are discussed.


Assuntos
GMP Cíclico , Modelos Animais de Doenças , Retina/efeitos dos fármacos , Degeneração Retiniana/genética , Animais , GMP Cíclico/genética , GMP Cíclico/metabolismo , GMP Cíclico/toxicidade , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Humanos , Camundongos , Camundongos Mutantes , Mutação/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Visão Ocular/genética , Peixe-Zebra
3.
Res Sq ; 2023 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-37790324

RESUMO

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes. This regeneration requires Müller glia (MG) to reprogram and divide asymmetrically to produce a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, does loss of different retinal cell subtypes induce unique MG regeneration responses? Second, do MG reprogram to a developmental retinal progenitor cell state? And finally, to what extent does regeneration recapitulate retinal development? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. While MG reprogram to a state similar to late-stage retinal progenitors in developing retinas, there are transcriptional differences between reprogrammed MG/MGPCs and late progenitors, as well as reprogrammed MG in outer and inner retinal damage models. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes. This work identifies major differences between gene regulatory networks activated following the selective loss of different subtypes of retina neurons, as well as between retinal regeneration and development.

4.
bioRxiv ; 2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37609307

RESUMO

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes. This regeneration requires Müller glia (MG) to reprogram and divide asymmetrically to produce a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, does loss of different retinal cell subtypes induce unique MG regeneration responses? Second, do MG reprogram to a developmental retinal progenitor cell state? And finally, to what extent does regeneration recapitulate retinal development? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. While MG reprogram to a state similar to late-stage retinal progenitors in developing retinas, there are transcriptional differences between reprogrammed MG/MGPCs and late progenitors, as well as reprogrammed MG in outer and inner retinal damage models. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes. This work identifies major differences between gene regulatory networks activated following the selective loss of different subtypes of retina neurons, as well as between retinal regeneration and development.

5.
Nat Commun ; 14(1): 8477, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38123561

RESUMO

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes through Müller glia (MG) reprogramming and asymmetric cell division that produces a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, do MG reprogram to a developmental retinal progenitor cell (RPC) state? Second, to what extent does regeneration recapitulate retinal development? And finally, does loss of different retinal cell subtypes induce unique MG regeneration responses? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. Here we show that injury induces MG to reprogram to a state similar to late-stage RPCs. However, there are major transcriptional differences between MGPCs and RPCs, as well as major transcriptional differences between activated MG and MGPCs when different retinal cell subtypes are damaged. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes.


Assuntos
Redes Reguladoras de Genes , Peixe-Zebra , Animais , Peixe-Zebra/genética , Retina/metabolismo , Neurogênese/genética , Neuroglia/metabolismo , Proliferação de Células/fisiologia , Células Ependimogliais/metabolismo
6.
Front Cell Dev Biol ; 10: 892271, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36120571

RESUMO

Unlike mammals, zebrafish regenerate in response to retinal damage. Because microglia are activated by retinal damage, we investigated their role during regeneration following either acute or chronic damage. At three weeks post-fertilization (wpf), both wild-type fish exhibiting NMDA-induced acute ganglion and amacrine cell death and gold rush (gosh) mutant fish possessing chronic cone photoreceptor degeneration displayed reactive microglia/macrophages and Müller glia proliferation. Dexamethasone-treated retinas, to inhibit the immune response, lacked reactive microglia/macrophages and possessed fewer PCNA-positive cells, while LPS treatment increased microglia/macrophages and PCNA-labeled cells. NMDA-injured retinas upregulated expression of il-1ß and tnfα pro-inflammatory cytokine genes, followed by increased expression of il-10 and arg1 anti-inflammatory/remodeling cytokine genes. A transient early TNFα pro-inflammatory microglia/macrophage population was visualized in NMDA-damaged retinas. In contrast, gosh mutant retinas exhibited a slight increase of pro-inflammatory cytokine gene expression concurrently with a greater increased anti-inflammatory/remodeling cytokine gene expression. Few TNFα pro-inflammatory microglia/macrophages were observed in the gosh retina. Understanding why acute and chronic damage results in different inflammation profiles and their effects on regulating zebrafish retinal regeneration would provide important clues toward improving therapeutic strategies for repairing injured mammalian tissues.

7.
Neural Regen Res ; 16(9): 1693-1701, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33510057

RESUMO

Tissue or organ regeneration is a complex process with successful outcomes depending on the type of tissue and organism. Upon damage, mammals can only efficiently restore a few tissues including the liver, skin, epithelia of the lung, kidney, and gut. In contrast, lower vertebrates such as zebrafish possess an extraordinary regeneration ability, which restores the normal function of a broad spectrum of tissues including heart, fin, brain, spinal cord, and retina. This regeneration process is either mediated by the proliferation of resident stem cells, or cells that dedifferentiate into a stem cell-like. In recent years, evidence has suggested that the innate immune system can modulate stem cell activity to initiate the regenerative response to damage. This review will explore some of the newer concepts of inflammation in zebrafish regeneration in different tissues. Understanding how inflammation regulates regeneration in zebrafish would provide important clues to improve the therapeutic strategies for repairing injured mammalian tissues that do not have an inherent regenerative capacity.

9.
Front Cell Dev Biol ; 7: 296, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31998714

RESUMO

Unlike mammals, zebrafish have the capacity to regenerate neurons in response to damage. Most zebrafish retinal injury models employ acute damage, which is unlike the chronic, gradual damage that occurs in human retinal diseases. Here, we studied the regenerative response in the zebrafish aipl1b mutant, gold rush (gosh). In gosh mutants, both cones and rods degenerate by 3 weeks post-fertilization (wpf). Müller glia do not exhibit a regenerative response by 3 wpf; however, they do present non-proliferative gliosis. Only at 5 wpf, is proliferation of Müller cells and rod precursor cells activated. Rods start to recover at 5 wpf and by 12 wpf they reach a level of recovery comparable to wild type, but cones remain absent in the adult stage. TNFα was detected in degenerating cones at 5-7 wpf and in Müller glia at 7 wpf in gosh mutants. At 5 wpf, proliferating Müller glia express Sox2, followed by Pax6 expression in neuronal progenitor cells (NPCs), confirming that the neuronal regeneration program is activated in gosh mutants after 5 wpf. Although acute light-induced damage did not activate proliferation of Müller glia, TNFα injection caused Müller glia to commence a proliferative response at 3 wpf in gosh mutants. These results suggest that Müller glia transition from non-proliferative gliosis to a regenerative state in gosh mutants, and that ectopic introduction of TNFα promotes this Müller cell transition even at 3 wpf. Thus, zebrafish gosh mutants provide a useful model to investigate mechanisms underlying retinal regeneration in a chronic photoreceptor degeneration model.

10.
Invest Ophthalmol Vis Sci ; 48(3): 1348-54, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17325183

RESUMO

PURPOSE: Damage induced by detachment of the neural retina and the retinal pigment epithelium (RPE) can be reduced by dark adaptation. The authors evaluated the influence of the duration of dark adaptation, time of day, and modification of the melatonin-dopamine pathway on acute RPE lesions induced by mechanical detachment. METHODS: BALB/c mice were studied at different times of day and different periods of dark adaptation. Some mice were treated with melatonin or sulpiride, a D2 dopamine receptor antagonist. Enucleated eyes and different saline solutions were used in experiments ex vivo. Retinal detachments in vivo were made by subretinal injections of hyaluronic acid. RPE cell damage was quantitatively evaluated with a dye exclusion procedure, and their viability was tested by preservation of tight junctions in culture. Lectin histochemistry was used to examine the interphotoreceptor matrix (IPM). RESULTS: Significant propidium iodide (PI) incorporation in RPE cells was detected after ex vivo separation during daytime, but it was very low when detachment took place at night after 24 to 48 hours of dark adaptation. PI exclusion was achieved during daytime after a single hour of dark adaptation when mice were pretreated with melatonin or sulpiride. Reduction of RPE cell damage was accompanied by decreased lectin binding to cone sheaths. CONCLUSIONS: A combination of time of day and length of dark adaptation decreased damage induced by detachment of the retina ex vivo and in vivo. Melatonin or sulpiride could replace these environmental factors. Therefore, melatonin and dopamine pathways might be involved in the control of IPM properties and retina/RPE interactions.


Assuntos
Epitélio Pigmentado Ocular/patologia , Descolamento Retiniano/patologia , Animais , Sobrevivência Celular , Células Cultivadas , Adaptação à Escuridão , Antagonistas dos Receptores de Dopamina D2 , Matriz Extracelular , Histocitoquímica , Lectinas/metabolismo , Masculino , Melatonina/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Modelos Animais , Fosfoproteínas/metabolismo , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/metabolismo , Propídio/metabolismo , Sulpirida/farmacologia , Fatores de Tempo , Proteína da Zônula de Oclusão-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA