Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Commun ; 9(1): 2389, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921864

RESUMO

Early during PNS regeneration, regenerating axons emerge from the proximal nerve stump, yet whether they extend simultaneously or whether pioneering axons establish a path for follower axons remains unknown. Moreover, the molecular mechanisms underlying robust regeneration are incompletely understood. Using live imaging, we demonstrate that in zebrafish pioneering axons establish a regenerative path for follower axons. We find this process requires the synaptic receptor lrp4, and in lrp4 mutants pioneers are unaffected while follower axons frequently stall at the injury gap, providing evidence for molecular diversity between pioneering and follower axons in regeneration. We demonstrate that Lrp4 promotes regeneration through an axon extrinsic mechanism and independent of membrane anchoring and MuSK co-receptor signaling essential for synaptic development. Finally, we show that Lrp4 coordinates the realignment of denervated Schwann cells with regenerating axons, consistent with a model by which Lrp4 is repurposed to promote sustained peripheral nerve regeneration via axon-glia interactions.


Assuntos
Proteínas Relacionadas a Receptor de LDL/metabolismo , Regeneração Nervosa , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Axônios/fisiologia , Proteínas de Homeodomínio , Proteínas Relacionadas a Receptor de LDL/genética , Microscopia Confocal , Mutação , Neuroglia/metabolismo , Neuroglia/fisiologia , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/fisiopatologia , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Células de Schwann/metabolismo , Células de Schwann/fisiologia , Imagem com Lapso de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
2.
Proc Natl Acad Sci U S A ; 112(9): 2859-64, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25691753

RESUMO

Following their synthesis in the endoplasmic reticulum (ER), voltage-gated sodium channels (NaV) are transported to the membranes of excitable cells, where they often cluster, such as at the axon initial segment of neurons. Although the mechanisms by which NaV channels form and maintain clusters have been extensively examined, the processes that govern their transport and degradation have received less attention. Our entry into the study of these processes began with the isolation of a new allele of the zebrafish mutant alligator, which we found to be caused by mutations in the gene encoding really interesting new gene (RING) finger protein 121 (RNF121), an E3-ubiquitin ligase present in the ER and cis-Golgi compartments. Here we demonstrate that RNF121 facilitates two opposing fates of NaV channels: (i) ubiquitin-mediated proteasome degradation and (ii) membrane localization when coexpressed with auxiliary NaVß subunits. Collectively, these results indicate that RNF121 participates in the quality control of NaV channels during their synthesis and subsequent transport to the membrane.


Assuntos
Proteólise , Domínios RING Finger , Ubiquitina-Proteína Ligases/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Sequência de Bases , Membrana Celular/genética , Membrana Celular/metabolismo , Dados de Sequência Molecular , Mutação , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico/fisiologia , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Canais de Sódio Disparados por Voltagem/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
J Neurosci ; 34(29): 9644-55, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25031404

RESUMO

Spontaneous network activity is a highly stereotyped early feature of developing circuits throughout the nervous system, including in the spinal cord. Spinal locomotor circuits produce a series of behaviors during development before locomotion that reflect the continual integration of spinal neurons into a functional network, but how the circuitry is reconfigured is not understood. The first behavior of the zebrafish embryo (spontaneous coiling) is mediated by an electrical circuit that subsequently generates mature locomotion (swimming) as chemical neurotransmission develops. We describe here a new spontaneous behavior, double coiling, that consists of two alternating contractions of the tail in rapid succession. Double coiling was glutamate-dependent and required descending hindbrain excitation, similar to but preceding swimming, making it a discrete intermediary developmental behavior. At the cellular level, motoneurons had a distinctive glutamate-dependent activity pattern that correlated with double coiling. Two glutamatergic interneurons, CoPAs and CiDs, had different activity profiles during this novel behavior. CoPA neurons failed to show changes in activity patterns during the period in which double coiling appears, whereas CiD neurons developed a glutamate-dependent activity pattern that correlated with double coiling and they innervated motoneurons at that time. Additionally, double coils were modified after pharmacological reduction of glycinergic neurotransmission such that embryos produced three or more rapidly alternating coils. We propose that double coiling behavior represents an important transition of the motor network from an electrically coupled spinal cord circuit that produces simple periodic coils to a spinal network driven by descending chemical neurotransmission, which generates more complex behaviors.


Assuntos
Atividade Motora/fisiologia , Neurônios Motores/fisiologia , Rede Nervosa/fisiologia , Medula Espinal/citologia , Sinapses/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Estimulação Elétrica , Embrião não Mamífero , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Atividade Motora/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Vias Neurais/embriologia , Rombencéfalo/fisiologia , Medula Espinal/embriologia , Sinapses/classificação , Sinapses/efeitos dos fármacos , Fatores de Transcrição/genética , Valina/análogos & derivados , Valina/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
4.
Nat Commun ; 4: 1952, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23736855

RESUMO

Excitation-contraction coupling, the process that regulates contractions by skeletal muscles, transduces changes in membrane voltage by activating release of Ca(2+) from internal stores to initiate muscle contraction. Defects in excitation-contraction coupling are associated with muscle diseases. Here we identify Stac3 as a novel component of the excitation-contraction coupling machinery. Using a zebrafish genetic screen, we generate a locomotor mutation that is mapped to stac3. We provide electrophysiological, Ca(2+) imaging, immunocytochemical and biochemical evidence that Stac3 participates in excitation-contraction coupling in muscles. Furthermore, we reveal that a mutation in human STAC3 is the genetic basis of the debilitating Native American myopathy (NAM). Analysis of NAM stac3 in zebrafish shows that the NAM mutation decreases excitation-contraction coupling. These findings enhance our understanding of both excitation-contraction coupling and the pathology of myopathies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Fissura Palatina/genética , Fissura Palatina/fisiopatologia , Acoplamento Excitação-Contração , Hipertermia Maligna/genética , Hipertermia Maligna/fisiopatologia , Mutação/genética , Miotonia Congênita/genética , Miotonia Congênita/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas de Peixe-Zebra/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Embrião não Mamífero/metabolismo , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Miofibrilas/metabolismo , Miofibrilas/ultraestrutura , Miotonia Congênita/patologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Especificidade de Órgãos/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Natação , Tato , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo
5.
J Neurosci ; 33(9): 3834-43, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447595

RESUMO

Nonvisual photosensation enables animals to sense light without sight. However, the cellular and molecular mechanisms of nonvisual photobehaviors are poorly understood, especially in vertebrate animals. Here, we describe the photomotor response (PMR), a robust and reproducible series of motor behaviors in zebrafish that is elicited by visual wavelengths of light but does not require the eyes, pineal gland, or other canonical deep-brain photoreceptive organs. Unlike the relatively slow effects of canonical nonvisual pathways, motor circuits are strongly and quickly (seconds) recruited during the PMR behavior. We find that the hindbrain is both necessary and sufficient to drive these behaviors. Using in vivo calcium imaging, we identify a discrete set of neurons within the hindbrain whose responses to light mirror the PMR behavior. Pharmacological inhibition of the visual cycle blocks PMR behaviors, suggesting that opsin-based photoreceptors control this behavior. These data represent the first known light-sensing circuit in the vertebrate hindbrain.


Assuntos
Movimento/fisiologia , Opsinas/metabolismo , Células Fotorreceptoras de Vertebrados/fisiologia , Rombencéfalo/citologia , Comportamento Estereotipado/fisiologia , Fatores Etários , Análise de Variância , Animais , Fenômenos Biomecânicos , Biofísica , Cálcio/metabolismo , Embrião não Mamífero , Feminino , Masculino , Microscopia Confocal , Morfolinos/farmacologia , Movimento/efeitos dos fármacos , Movimento/efeitos da radiação , Células Musculares/efeitos dos fármacos , Células Musculares/efeitos da radiação , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Vias Neurais/efeitos da radiação , Opsinas/química , Estimulação Luminosa , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Rombencéfalo/fisiologia , Comportamento Estereotipado/efeitos dos fármacos , Comportamento Estereotipado/efeitos da radiação , Fatores de Tempo , Peixe-Zebra
6.
J Neurophysiol ; 108(1): 148-59, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22490555

RESUMO

The molecular and physiological basis of the touch-unresponsive zebrafish mutant fakir has remained elusive. Here we report that the fakir phenotype is caused by a missense mutation in the gene encoding voltage-gated calcium channel 2.1b (CACNA1Ab). Injection of RNA encoding wild-type CaV2.1 restores touch responsiveness in fakir mutants, whereas knockdown of CACNA1Ab via morpholino oligonucleotides recapitulates the fakir mutant phenotype. Fakir mutants display normal current-evoked synaptic communication at the neuromuscular junction but have attenuated touch-evoked activation of motor neurons. NMDA-evoked fictive swimming is not affected by the loss of CaV2.1b, suggesting that this channel is not required for motor pattern generation. These results, coupled with the expression of CACNA1Ab by sensory neurons, suggest that CaV2.1b channel activity is necessary for touch-evoked activation of the locomotor network in zebrafish.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Ativação do Canal Iônico/genética , Tato/genética , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Vias Aferentes/fisiologia , Animais , Animais Geneticamente Modificados , Bungarotoxinas/metabolismo , Canais de Cálcio Tipo N/genética , Curare/farmacologia , Relação Dose-Resposta a Droga , Embrião não Mamífero , Reação de Fuga/efeitos dos fármacos , Reação de Fuga/fisiologia , Potenciais Evocados/genética , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Leucina/genética , Locomoção/efeitos dos fármacos , Locomoção/genética , Modelos Moleculares , Morfolinas/farmacologia , Atividade Motora/genética , Neurônios Motores/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Mutagênese Sítio-Dirigida/métodos , Mutação/genética , Mutação de Sentido Incorreto/genética , Rede Nervosa/fisiologia , Antagonistas Nicotínicos/farmacologia , Medula Espinal/citologia , Medula Espinal/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Tato/fisiologia , Valina/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
J Biol Chem ; 287(2): 1080-9, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22075003

RESUMO

In many tissues and organs, connexin proteins assemble between neighboring cells to form gap junctions. These gap junctions facilitate direct intercellular communication between adjoining cells, allowing for the transmission of both chemical and electrical signals. In rodents, gap junctions are found in differentiating myoblasts and are important for myogenesis. Although gap junctions were once believed to be absent from differentiated skeletal muscle in mammals, recent studies in teleosts revealed that differentiated muscle does express connexins and is electrically coupled, at least at the larval stage. These findings raised questions regarding the functional significance of gap junctions in differentiated muscle. Our analysis of gap junctions in muscle began with the isolation of a zebrafish motor mutant that displayed weak coiling at day 1 of development, a behavior known to be driven by slow-twitch muscle (slow muscle). We identified a missense mutation in the gene encoding Connexin 39.9. In situ hybridization found connexin 39.9 to be expressed by slow muscle. Paired muscle recordings uncovered that wild-type slow muscles are electrically coupled, whereas mutant slow muscles are not. The further examination of cellular activity revealed aberrant, arrhythmic touch-evoked Ca(2+) transients in mutant slow muscle and a reduction in the number of muscle fibers contracting in response to touch in mutants. These results indicate that Connexin 39.9 facilitates the spreading of neuronal inputs, which is irregular during motor development, beyond the muscle cells and that gap junctions play an essential role in the efficient recruitment of slow muscle fibers.


Assuntos
Conexinas/metabolismo , Proteínas Musculares/metabolismo , Mutação de Sentido Incorreto , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Sequência de Bases , Conexinas/genética , Junções Comunicantes/genética , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica/genética , Dados de Sequência Molecular , Fibras Musculares de Contração Lenta , Proteínas Musculares/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
J Neurosci ; 31(32): 11633-44, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21832193

RESUMO

Mutations in the gene encoding TRPM7 (trpm7), a member of the Transient Receptor Potential (TRP) superfamily of cation channels that possesses an enzymatically active kinase at its C terminus, cause the touch-unresponsive zebrafish mutant touchdown. We identified and characterized a new allele of touchdown, as well as two previously reported alleles, and found that all three alleles harbor mutations that abolish channel activity. Through the selective restoration of TRPM7 expression in sensory neurons, we found that TRPM7's kinase activity and selectivity for divalent cations over monovalent cations were dispensable for touch-evoked activation of escape behaviors in zebrafish. Additional characterization revealed that sensory neurons were present and capable of responding to tactile stimuli in touchdown mutants, indicating that TRPM7 is not required for sensory neuron survival or mechanosensation. Finally, exposure to elevated concentrations of divalent cations was found to restore touch-evoked behaviors in touchdown mutants. Collectively, these findings are consistent with a role for zebrafish TRPM7 within sensory neurons in the modulation of neurotransmitter release at central synapses, similar to that proposed for mammalian TRPM7 at peripheral synapses.


Assuntos
Alelos , Reação de Fuga/fisiologia , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPM/fisiologia , Tato/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Feminino , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/fisiologia , Proteínas Serina-Treonina Quinases , Especificidade da Espécie , Canais de Cátion TRPM/genética , Tato/genética , Xenopus , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
9.
Brain ; 134(Pt 2): 602-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21115467

RESUMO

Autosomal dominant sensory ataxia is a rare genetic condition that results in a progressive ataxia that is caused by degeneration of the posterior columns of the spinal cord. To date only two families have been clinically ascertained with this condition, both from Maritime Canada. We previously mapped both families to chromosome 8p12-8q12 and have now screened the majority of annotated protein-coding genes in the shared haplotype region by direct DNA sequencing. We have identified a putative pathogenic mutation in the gene encoding ring-finger protein RNF170, a potential ubiquitin ligase. This mutation is a rare non-synonymous change in a well-conserved residue and is predicted to be pathogenic by SIFT, PolyPhen, PANTHER and Align-GVD. Microinjection of wild-type or mutant orthologous messenger RNAs into zebrafish (Danio rerio) embryos confirmed that the mutation dominantly disrupts normal embryonic development. Together these results suggest that the mutation in RNF170 is causal for the sensory ataxia in these families.


Assuntos
Ataxia/genética , Mutação de Sentido Incorreto , Ubiquitina-Proteína Ligases/genética , Sequência de Aminoácidos , Animais , Ataxia/metabolismo , Morte Celular/efeitos dos fármacos , Variações do Número de Cópias de DNA , Técnicas de Silenciamento de Genes/métodos , Humanos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
10.
Prog Brain Res ; 187: 47-61, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21111200

RESUMO

The nervous system can generate rhythms of various frequencies; on the low-frequency side, we have the circuits regulating circadian rhythms with a 24-h period, while on the high-frequency side we have the motor circuits that underlie flight in a hummingbird. Given the ubiquitous nature of rhythms, it is surprising that we know very little of the cellular and molecular mechanisms that produce them in the embryos and of their potential role during the development of neuronal circuits. Recently, zebrafish has been developed as a vertebrate model to study the genetics of neural development. Zebrafish offer several advantages to the study of nervous system development including optical and electrophysiological analysis of neuronal activity even at the earliest embryonic stages. This unique combination of physiology and genetics in the same animal model has led to insights into the development of neuronal networks. This chapter reviews work on the development of zebrafish motor rhythms and speculates on birth and maturation of the circuits that produce them.


Assuntos
Atividade Motora/fisiologia , Periodicidade , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero/fisiologia , Mutação , Medula Espinal/anatomia & histologia , Medula Espinal/fisiologia
11.
J Neurosci ; 30(28): 9359-67, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20631165

RESUMO

The process by which light touch in vertebrates is transformed into an electrical response in cutaneous mechanosensitive neurons is a largely unresolved question. To address this question we undertook a forward genetic screen in zebrafish (Danio rerio) to identify mutants exhibiting abnormal touch-evoked behaviors, despite the presence of sensory neurons and peripheral neurites. One family, subsequently named touché, was found to harbor a recessive mutation which produced offspring that were unresponsive to light touch, but responded to a variety of other sensory stimuli. The optogenetic activation of motor behaviors by touché mutant sensory neurons expressing channelrhodopsin-2 suggested that the synaptic output of sensory neurons was intact, consistent with a defect in sensory neuron activation. To explore sensory neuron activation we developed an in vivo preparation permitting the precise placement of a combined electrical and tactile stimulating probe upon eGFP-positive peripheral neurites. In wild-type larva electrical and tactile stimulation of peripheral neurites produced action potentials detectable within the cell body. In a subset of these sensory neurons an underlying generator potential could be observed in response to subthreshold tactile stimuli. A closer examination revealed that the amplitude of the generator potential was proportional to the stimulus amplitude. When assayed touché mutant sensory neurons also responded to electrical stimulation of peripheral neurites similar to wild-type larvae, however tactile stimulation of these neurites failed to uncover a subset of sensory neurons possessing generator potentials. These findings suggest that touché is required for generator potentials, and that cutaneous mechanoreceptors with generator potentials are necessary for responsiveness to light touch in zebrafish.


Assuntos
Potenciais Somatossensoriais Evocados/fisiologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia , Tato/fisiologia , Proteínas de Peixe-Zebra/genética , Animais , Eletrofisiologia , Rede Nervosa/fisiologia , Neurônios Aferentes/fisiologia , Estimulação Física , Peixe-Zebra/genética
12.
Development ; 137(10): 1689-98, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20392743

RESUMO

In zebrafish, Rohon-Beard (RB) neurons are primary sensory neurons present during the embryonic and early larval stages. At 2 days post-fertilization (dpf), wild-type zebrafish embryos respond to mechanosensory stimulation and swim away from the stimuli, whereas mi310 mutants are insensitive to touch. During approximately 2-4 dpf, wild-type RB neurons undergo programmed cell death, which is caused by sodium current-mediated electrical activity, whereas mutant RB cells survive past 4 dpf, suggesting a defect of sodium currents in the mutants. Indeed, electrophysiological recordings demonstrated the generation of action potentials in wild-type RB neurons, whereas mutant RB cells failed to fire owing to the reduction of voltage-gated sodium currents. Labeling of dissociated RB neurons with an antibody against voltage-gated sodium channels revealed that sodium channels are expressed at the cell surface in wild-type, but not mutant, RB neurons. Finally, in mi310 mutants, we identified a mis-sense mutation in pigu, a subunit of GPI (glycosylphosphatidylinositol) transamidase, which is essential for membrane anchoring of GPI-anchored proteins. Taken together, biogenesis of GPI-anchored proteins is necessary for cell surface expression of sodium channels and thus for firings of RB neurons, which enable zebrafish embryos to respond to mechanosensory stimulation.


Assuntos
Glicosilfosfatidilinositóis/metabolismo , Mecanotransdução Celular/genética , Proteínas de Membrana/biossíntese , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/metabolismo , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Antígenos de Superfície/metabolismo , Células CHO , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Morte Celular/fisiologia , Cricetinae , Cricetulus , Embrião não Mamífero , Técnicas de Silenciamento de Genes , Glicosilfosfatidilinositóis/biossíntese , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Estimulação Física , RNA Interferente Pequeno/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
13.
Dev Neurobiol ; 70(7): 508-22, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20225246

RESUMO

A screen for zebrafish motor mutants identified two noncomplementing alleles of a recessive mutation that were named non-active (nav(mi89) and nav(mi130)). nav embryos displayed diminished spontaneous and touch-evoked escape behaviors during the first 3 days of development. Genetic mapping identified the gene encoding Na(V)1.6a (scn8aa) as a potential candidate for nav. Subsequent cloning of scn8aa from the two alleles of nav uncovered two missense mutations in Na(V)1.6a that eliminated channel activity when assayed heterologously. Furthermore, the injection of RNA encoding wild-type scn8aa rescued the nav mutant phenotype indicating that scn8aa was the causative gene of nav. In-vivo electrophysiological analysis of the touch-evoked escape circuit indicated that voltage-dependent inward current was decreased in mechanosensory neurons in mutants, but they were able to fire action potentials. Furthermore, tactile stimulation of mutants activated some neurons downstream of mechanosensory neurons but failed to activate the swim locomotor circuit in accord with the behavioral response of initial escape contractions but no swimming. Thus, mutant mechanosensory neurons appeared to respond to tactile stimulation but failed to initiate swimming. Interestingly fictive swimming could be initiated pharmacologically suggesting that a swim circuit was present in mutants. These results suggested that Na(V)1.6a was required for touch-induced activation of the swim locomotor network.


Assuntos
Sistema Nervoso Central/embriologia , Rede Nervosa/embriologia , Canais de Sódio/fisiologia , Natação/fisiologia , Tato/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Comportamento Animal/fisiologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Mapeamento Cromossômico , Feminino , Masculino , Mecanorreceptores/citologia , Mecanorreceptores/metabolismo , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto/genética , Canal de Sódio Disparado por Voltagem NAV1.6 , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Canais de Sódio/genética , Tato/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
14.
Dev Neurobiol ; 69(12): 780-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19634126

RESUMO

Characterizing connectivity in the spinal cord of zebrafish embryos is not only prerequisite to understanding the development of locomotion, but is also necessary for maximizing the potential of genetic studies of circuit formation in this model system. During their first day of development, zebrafish embryos show two simple motor behaviors. First, they coil their trunks spontaneously, and a few hours later they start responding to touch with contralateral coils. These behaviors are contemporaneous until spontaneous coils become infrequent by 30 h. Glutamatergic neurons are distributed throughout the embryonic spinal cord, but their contribution to these early motor behaviors in immature zebrafish is still unclear. We demonstrate that the kinetics of spontaneous coiling and touch-evoked responses show distinct developmental time courses and that the touch response is dependent on AMPA-type glutamate receptor activation. Transection experiments suggest that the circuits required for touch-evoked responses are confined to the spinal cord and that only the most rostral part of the spinal cord is sufficient for triggering the full response. This rostral sensory connection is presumably established via CoPA interneurons, as they project to the rostral spinal cord. Electrophysiological analysis demonstrates that these neurons receive short latency AMPA-type glutamatergic inputs in response to ipsilateral tactile stimuli. We conclude that touch responses in early embryonic zebrafish arise only after glutamatergic synapses connect sensory neurons and interneurons to the contralateral motor network via a rostral loop. This helps define an elementary circuit that is modified by the addition of sensory inputs, resulting in behavioral transformation.


Assuntos
Reação de Fuga/fisiologia , Ácido Glutâmico/metabolismo , Receptores de AMPA/metabolismo , Reflexo/fisiologia , Medula Espinal/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Maleato de Dizocilpina/farmacologia , Eletrofisiologia , Embrião não Mamífero , Reação de Fuga/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Imuno-Histoquímica , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Reflexo/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/embriologia , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Natação/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/fisiologia , Peixe-Zebra
15.
Nat Genet ; 41(6): 651-3, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19412178

RESUMO

The sideroblastic anemias are a heterogeneous group of congenital and acquired hematological disorders whose morphological hallmark is the presence of ringed sideroblasts--bone marrow erythroid precursors containing pathologic iron deposits within mitochondria. Here, by positional cloning, we define a previously unknown form of autosomal recessive nonsyndromic congenital sideroblastic anemia, associated with mutations in the gene encoding the erythroid specific mitochondrial carrier family protein SLC25A38, and demonstrate that SLC25A38 is important for the biosynthesis of heme in eukaryotes.


Assuntos
Anemia Sideroblástica/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Mutação , Animais , Portador Sadio , Família , Peixes/genética , Heme/biossíntese , Humanos , Fenótipo , Leveduras/genética
16.
Dev Neurobiol ; 68(1): 45-61, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17918238

RESUMO

The zebrafish ennui mutation was identified from a mutagenesis screen for defects in early behavior. Homozygous ennui embryos swam more slowly than wild-type siblings but normal swimming recovered during larval stages and homozygous mutants survived until adulthood. Electrophysiological recordings from motoneurons and muscles suggested that the motor output of the CNS following mechanosensory stimulation was normal in ennui, but the synaptic currents at the neuromuscular junction were significantly reduced. Analysis of acetylcholine receptors (AChRs) in ennui muscles showed a marked reduction in the size of synaptic clusters and their aberrant localization at the myotome segment borders of fast twitch muscle. Prepatterned, nerve-independent AChR clusters appeared normal in mutant embryos and dispersed upon outgrowth of motor axons onto the muscles. Genetic mosaic analysis showed that ennui is required cell autonomously in muscle fibers for normal synaptic localization of AChRs. Furthermore, exogenous agrin failed to induce AChR aggregation, suggesting that ennui is crucial for agrin function. Finally, motor axons branched more extensively in ennui fast twitch muscles especially in the region of the myotome borders. These results suggest that ennui is important for nerve-dependent AChR clustering and the stability of axon growth.


Assuntos
Axônios/fisiologia , Neurônios Motores/patologia , Transtornos dos Movimentos , Mutação/fisiologia , Receptores Colinérgicos/metabolismo , Proteínas de Peixe-Zebra/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Agrina/farmacologia , Animais , Axônios/efeitos dos fármacos , Comportamento Animal , Embrião não Mamífero , Neurônios Motores/efeitos dos fármacos , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/patologia , Transtornos dos Movimentos/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/fisiopatologia , Transporte Proteico/efeitos dos fármacos , Natação , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
17.
Development ; 134(15): 2771-81, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17596281

RESUMO

Wild-type zebrafish embryos swim away in response to tactile stimulation. By contrast, relatively relaxed mutants swim slowly due to weak contractions of trunk muscles. Electrophysiological recordings from muscle showed that output from the CNS was normal in mutants, suggesting a defect in the muscle. Calcium imaging revealed that Ca(2+) transients were reduced in mutant fast muscle. Immunostaining demonstrated that ryanodine and dihydropyridine receptors, which are responsible for Ca(2+) release following membrane depolarization, were severely reduced at transverse-tubule/sarcoplasmic reticulum junctions in mutant fast muscle. Thus, slow swimming is caused by weak muscle contractions due to impaired excitation-contraction coupling. Indeed, most of the ryanodine receptor 1b (ryr1b) mRNA in mutants carried a nonsense mutation that was generated by aberrant splicing due to a DNA insertion in an intron of the ryr1b gene, leading to a hypomorphic condition in relatively relaxed mutants. RYR1 mutations in humans lead to a congenital myopathy, multi-minicore disease (MmD), which is defined by amorphous cores in muscle. Electron micrographs showed minicore structures in mutant fast muscles. Furthermore, following the introduction of antisense morpholino oligonucleotides that restored the normal splicing of ryr1b, swimming was recovered in mutants. These findings suggest that zebrafish relatively relaxed mutants may be useful for understanding the development and physiology of MmD.


Assuntos
Modelos Animais de Doenças , Doenças Musculares/genética , Doenças Musculares/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Natação , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Sinalização do Cálcio/fisiologia , Sistema Nervoso Central/fisiologia , Embrião não Mamífero , Modelos Biológicos , Dados de Sequência Molecular , Contração Muscular/genética , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Rápida/fisiologia , Doenças Musculares/congênito , Doenças Musculares/fisiopatologia , Isoformas de Proteínas/genética , Sítios de Splice de RNA/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Peixe-Zebra/embriologia
18.
Cell Calcium ; 39(3): 227-36, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16368137

RESUMO

Contractions by skeletal muscle require proper excitation-contraction (EC) coupling, whereby depolarization of the muscle membrane leads to an increase in cytosolic Ca(2+) and contraction. Changes in membrane voltage are detected by dihydropyridine receptors (DHPR) that directly interact with and activate ryanodine receptors to release Ca(2+) from the sarcoplasmic reticulum into the cytosol. A genetic screen for motility mutations isolated a new allele of the immotile zebrafish mutant relaxed. Muscles in relaxed embryos do not contract in response to potassium chloride (KCl) thus appear unresponsive to membrane depolarization, but do contract when stimulated by caffeine, an agonist of ryanodine receptors. This suggests that relaxed mutant muscles are defective in EC coupling. Indeed, immunohistochemical analysis demonstrated that mutants lack DHPRs in skeletal muscles. The mutant phenotype results from non-sense mutations in the zebrafish CACNB1 gene that encodes the DHPR beta1 subunit. The zebrafish CACNB1 gene is expressed in skeletal muscles, PNS and CNS. Electrophysiological recordings showed no obvious abnormalities in the motor output of relaxed mutants, presumably due to redundancy provided by other beta subunits. The structural and functional homology of CACNB1 in zebrafish and mammals, suggests that zebrafish can be useful for studying EC coupling and potential neuronal function of CACNB1.


Assuntos
Canais de Cálcio Tipo L/genética , Códon sem Sentido , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Proteínas de Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Dados de Sequência Molecular , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Sistema Nervoso/metabolismo , Paralisia/genética , Alinhamento de Sequência , Peixe-Zebra/genética
19.
Zebrafish ; 3(2): 173-90, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-18248259

RESUMO

General mechanisms of motor network development have often been examined in the spinal cord because of its relative simplicity when compared to higher parts of the brain. Indeed, most of our current understanding of motor pattern generation comes from work in the lower vertebrate spinal cord. Nevertheless, very little is known about the initial stages of motor network formation and the interplay between genes and electrical activity. Recent research has led to the establishment of the zebrafish as a key model system to study the genetics of neural development. The spinal cord of zebrafish is amenable to optical and electrophysiological analysis of neuronal activity even at the earliest embryonic stages when the network is immature. The combination of physiology and genetics in the same animal model should lead to insights into the basic mechanisms of motor circuit formation. This paper reviews recent work on the development of zebrafish motor activity and discusses them in the context of the current knowledge of embryonic and larval zebrafish spinal cord morphology and physiology.

20.
J Neurosci ; 25(28): 6610-20, 2005 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-16014722

RESUMO

shocked (sho) is a zebrafish mutation that causes motor deficits attributable to CNS defects during the first2dof development. Mutant embryos display reduced spontaneous coiling of the trunk, diminished escape responses when touched, and an absence of swimming. A missense mutation in the slc6a9 gene that encodes a glycine transporter (GlyT1) was identified as the cause of the sho phenotype. Antisense knock-down of GlyT1 in wild-type embryos phenocopies sho, and injection of wild-type GlyT1 mRNA into mutants rescues them. A comparison of glycine-evoked inward currents in Xenopus oocytes expressing either the wild-type or mutant protein found that the missense mutation results in a nonfunctional transporter. glyt1 and the related glyt2 mRNAs are expressed in the hindbrain and spinal cord in nonoverlapping patterns. The fact that these regions are known to be required for generation of early locomotory behaviors suggests that the regulation of extracellular glycine levels in the CNS is important for proper function of neural networks. Furthermore, physiological analysis after manipulation of glycinergic activity in wild-type and sho embryos suggests that the mutant phenotype is attributable to elevated extracellular glycine within the CNS.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas da Membrana Plasmática de Transporte de Glicina/fisiologia , Glicina/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/genética , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/patologia , Embrião não Mamífero/fisiopatologia , Líquido Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Glicina/deficiência , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Músculos/embriologia , Músculos/fisiologia , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Oócitos , Fenótipo , Estimulação Física , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Sarcosina/análogos & derivados , Sarcosina/farmacologia , Medula Espinal/embriologia , Medula Espinal/metabolismo , Natação , Xenopus , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA