Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Biol ; 502: 1-13, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37290497

RESUMO

Zebrafish robustly regenerate fins, including their characteristic bony ray skeleton. Amputation activates intra-ray fibroblasts and dedifferentiates osteoblasts that migrate under a wound epidermis to establish an organized blastema. Coordinated proliferation and re-differentiation across lineages then sustains progressive outgrowth. We generate a single cell transcriptome dataset to characterize regenerative outgrowth and explore coordinated cell behaviors. We computationally identify sub-clusters representing most regenerative fin cell lineages, and define markers of osteoblasts, intra- and inter-ray fibroblasts and growth-promoting distal blastema cells. A pseudotemporal trajectory and in vivo photoconvertible lineage tracing indicate distal blastemal mesenchyme restores both intra- and inter-ray fibroblasts. Gene expression profiles across this trajectory suggest elevated protein production in the blastemal mesenchyme state. O-propargyl-puromycin incorporation and small molecule inhibition identify insulin growth factor receptor (IGFR)/mechanistic target of rapamycin kinase (mTOR)-dependent elevated bulk translation in blastemal mesenchyme and differentiating osteoblasts. We test candidate cooperating differentiation factors identified from the osteoblast trajectory, finding IGFR/mTOR signaling expedites glucocorticoid-promoted osteoblast differentiation in vitro. Concordantly, mTOR inhibition slows but does not prevent fin regenerative outgrowth in vivo. IGFR/mTOR may elevate translation in both fibroblast- and osteoblast-lineage cells during the outgrowth phase as a tempo-coordinating rheostat.


Assuntos
Transdução de Sinais , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Diferenciação Celular , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Receptores de Somatomedina/metabolismo , Nadadeiras de Animais/metabolismo
2.
Development ; 148(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34061172

RESUMO

Organs stop growing to achieve a characteristic size and shape in scale with the body of an animal. Likewise, regenerating organs sense injury extents to instruct appropriate replacement growth. Fish fins exemplify both phenomena through their tremendous diversity of form and remarkably robust regeneration. The classic zebrafish mutant longfint2 develops and regenerates dramatically elongated fins and underlying ray skeleton. We show longfint2 chromosome 2 overexpresses the ether-a-go-go-related voltage-gated potassium channel kcnh2a. Genetic disruption of kcnh2a in cis rescues longfint2, indicating longfint2 is a regulatory kcnh2a allele. We find longfint2 fin overgrowth originates from prolonged outgrowth periods by showing Kcnh2a chemical inhibition during late stage regeneration fully suppresses overgrowth. Cell transplantations demonstrate longfint2-ectopic kcnh2a acts tissue autonomously within the fin intra-ray mesenchymal lineage. Temporal inhibition of the Ca2+-dependent phosphatase calcineurin indicates it likewise entirely acts late in regeneration to attenuate fin outgrowth. Epistasis experiments suggest longfint2-expressed Kcnh2a inhibits calcineurin output to supersede growth cessation signals. We conclude ion signaling within the growth-determining mesenchyme lineage controls fin size by tuning outgrowth periods rather than altering positional information or cell-level growth potency.


Assuntos
Nadadeiras de Animais/fisiologia , Expressão Ectópica do Gene/fisiologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Nadadeiras de Animais/anatomia & histologia , Animais , Sistemas CRISPR-Cas , Calcineurina/metabolismo , Proliferação de Células , Expressão Ectópica do Gene/genética , Éter , Canais de Potássio Éter-A-Go-Go/genética , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Tamanho do Órgão , Regeneração/fisiologia , Transdução de Sinais/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
bioRxiv ; 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39229087

RESUMO

Adult zebrafish fins regenerate to their original size regardless of damage extent, providing a tractable model of organ size and scale control. Gain-of-function of voltage-gated K + channels expressed in fibroblast-lineage blastema cells promotes excessive fin outgrowth, leading to a long-finned phenotype. Similarly, inhibition of the Ca 2+ -dependent phosphatase calcineurin during regeneration causes dramatic fin overgrowth. However, Ca 2+ fluxes and their potential origins from dynamic membrane voltages have not been explored or linked to fin size restoration. We used fibroblast-lineage GCaMP imaging of regenerating adult fins to identify dynamic and heterogeneous Ca 2+ transients in distal blastema cells. Membrane depolarization of isolated regenerating fin fibroblasts triggered Ca 2+ spikes dependent on voltage-gated Ca 2+ channel activity. Single cell transcriptomics identified the voltage-gated Ca 2+ channels cacna1c (L-type channel), cacna1ba (N-type), and cacna1g (T-type) as candidate mediators of fibroblast-lineage Ca 2+ signaling. Small molecule inhibition revealed L- and/or N-type voltage-gated Ca 2+ channels act during regenerative outgrowth to restore fins to their original scale. Strikingly, cacna1g homozygous mutant zebrafish regenerated extraordinarily long fins due to prolonged outgrowth. The regenerated fins far exceeded their original length but with otherwise normal ray skeletons. Therefore, cacna1g mutants uniquely provide a genetic loss-of-function long-finned model that decouples developmental and regenerative fin outgrowth. Live GCaMP imaging of regenerating fins showed T-type Cacna1g channels enable Ca 2+ dynamics in distal fibroblast-lineage blastemal mesenchyme during the outgrowth phase. We conclude "bioelectricity" for fin size control likely entirely reflects voltage-modulated Ca 2+ dynamics in fibroblast-lineage blastemal cells that specifically and steadily decelerates outgrowth at a rate tuned to restore the original fin size.

4.
Neural Dev ; 2: 7, 2007 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-17470283

RESUMO

BACKGROUND: Proper neuronal function depends on forming three primary subcellular compartments: axons, dendrites, and soma. Each compartment has a specialized function (the axon to send information, dendrites to receive information, and the soma is where most cellular components are produced). In mammalian neurons, each primary compartment has distinctive molecular and morphological features, as well as smaller domains, such as the axon initial segment, that have more specialized functions. How neuronal subcellular compartments are established and maintained is not well understood. Genetic studies in Drosophila have provided insight into other areas of neurobiology, but it is not known whether flies are a good system in which to study neuronal polarity as a comprehensive analysis of Drosophila neuronal subcellular organization has not been performed. RESULTS: Here we use new and previously characterized markers to examine Drosophila neuronal compartments. We find that: axons and dendrites can accumulate different microtubule-binding proteins; protein synthesis machinery is concentrated in the cell body; pre- and post-synaptic sites localize to distinct regions of the neuron; and specializations similar to the initial segment are present. In addition, we track EB1-GFP dynamics and determine microtubules in axons and dendrites have opposite polarity. CONCLUSION: We conclude that Drosophila will be a powerful system to study the establishment and maintenance of neuronal compartments.


Assuntos
Encéfalo/citologia , Compartimento Celular/fisiologia , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Drosophila melanogaster/citologia , Neurônios/citologia , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Larva/citologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Modelos Animais , Corpos Pedunculados/citologia , Corpos Pedunculados/crescimento & desenvolvimento , Corpos Pedunculados/metabolismo , Neurônios/metabolismo , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA