Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Elife ; 112022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36222498

RESUMO

Development of elaborate and polarized neuronal morphology requires precisely regulated transport of cellular cargos by motor proteins such as kinesin-1. Kinesin-1 has numerous cellular cargos which must be delivered to unique neuronal compartments. The process by which this motor selectively transports and delivers cargo to regulate neuronal morphogenesis is poorly understood, although the cargo-binding kinesin light chain (KLC) subunits contribute to specificity. Our work implicates one such subunit, KLC4, as an essential regulator of axon branching and arborization pattern of sensory neurons during development. Using live imaging approaches in klc4 mutant zebrafish, we show that KLC4 is required for stabilization of nascent axon branches, proper microtubule (MT) dynamics, and endosomal transport. Furthermore, KLC4 is required for proper tiling of peripheral axon arbors: in klc4 mutants, peripheral axons showed abnormal fasciculation, a behavior characteristic of central axons. This result suggests that KLC4 patterns axonal compartments and helps establish molecular differences between central and peripheral axons. Finally, we find that klc4 mutant larva are hypersensitive to touch and adults show anxiety-like behavior in a novel tank test, implicating klc4 as a new gene involved in stress response circuits.


Assuntos
Cinesinas , Peixe-Zebra , Animais , Cinesinas/genética , Axônios/fisiologia , Células Receptoras Sensoriais/fisiologia , Morfogênese
2.
Dis Model Mech ; 15(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36037004

RESUMO

Neurofibromatosis type 1 (NF1) is an autosomal-dominant neurogenetic disorder caused by mutations in the gene neurofibromin 1 (NF1). NF1 predisposes individuals to a variety of symptoms, including peripheral nerve tumors, brain tumors and cognitive dysfunction. Cognitive deficits can negatively impact patient quality of life, especially the social and academic development of children. The neurofibromin protein influences neural circuits via diverse cellular signaling pathways, including through RAS, cAMP and dopamine signaling. Although animal models have been useful in identifying cellular and molecular mechanisms that regulate NF1-dependent behaviors, translating these discoveries into effective treatments has proven difficult. Clinical trials measuring cognitive outcomes in patients with NF1 have mainly targeted RAS signaling but, unfortunately, resulted in limited success. In this Review, we provide an overview of the structure and function of neurofibromin, and evaluate several cellular and molecular mechanisms underlying neurofibromin-dependent cognitive function, which have recently been delineated in animal models. A better understanding of neurofibromin roles in the development and function of the nervous system will be crucial for identifying new therapeutic targets for the various cognitive domains affected by NF1.


Assuntos
Disfunção Cognitiva , Neurofibromatose 1 , Animais , Genes da Neurofibromatose 1 , Modelos Animais , Neurofibromatose 1/complicações , Neurofibromatose 1/genética , Neurofibromina 1/genética , Qualidade de Vida
3.
Elife ; 102021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33759764

RESUMO

Endoplasmic reticulum (ER) and mitochondria form close physical associations to facilitate calcium transfer, thereby regulating mitochondrial function. Neurons with high metabolic demands, such as sensory hair cells, are especially dependent on precisely regulated ER-mitochondria associations. We previously showed that the secreted metalloprotease pregnancy-associated plasma protein-aa (Pappaa) regulates mitochondrial function in zebrafish lateral line hair cells (Alassaf et al., 2019). Here, we show that pappaa mutant hair cells exhibit excessive and abnormally close ER-mitochondria associations, suggesting increased ER-mitochondria calcium transfer. pappaa mutant hair cells are more vulnerable to pharmacological induction of ER-calcium transfer. Additionally, pappaa mutant hair cells display ER stress and dysfunctional downstream processes of the ER-mitochondria axis including altered mitochondrial morphology and reduced autophagy. We further show that Pappaa influences ER-calcium transfer and autophagy via its ability to stimulate insulin-like growth factor-1 bioavailability. Together our results identify Pappaa as a novel regulator of the ER-mitochondria axis.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Metaloendopeptidases/genética , Mitocôndrias/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Cálcio/metabolismo , Células Ciliadas Auditivas Internas/ultraestrutura , Sistema da Linha Lateral/ultraestrutura , Metaloendopeptidases/metabolismo , Microscopia Eletrônica de Transmissão e Varredura , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
4.
Mol Biol Cell ; 30(14): 1645-1654, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31091161

RESUMO

Mitotic spindles are well known to be assembled from and dependent on microtubules. In contrast, whether actin filaments (F-actin) are required for or are even present in mitotic spindles has long been controversial. Here we have developed improved methods for simultaneously preserving F-actin and microtubules in fixed samples and exploited them to demonstrate that F-actin is indeed associated with mitotic spindles in intact Xenopus laevis embryonic epithelia. We also find that there is an "F-actin cycle," in which the distribution and organization of spindle F-actin changes over the course of the cell cycle. Live imaging using a probe for F-actin reveals that at least two pools of F-actin are associated with mitotic spindles: a relatively stable internal network of cables that moves in concert with and appears to be linked to spindles, and F-actin "fingers" that rapidly extend from the cell cortex toward the spindle and make transient contact with the spindle poles. We conclude that there is a robust endoplasmic F-actin network in normal vertebrate epithelial cells and that this network is also a component of mitotic spindles. More broadly, we conclude that there is far more internal F-actin in epithelial cells than is commonly believed.


Assuntos
Actinas/metabolismo , Epitélio/metabolismo , Fuso Acromático/metabolismo , Xenopus laevis/metabolismo , Animais , Sobrevivência Celular , Retículo Endoplasmático/metabolismo , Células Epiteliais/metabolismo , Forminas/metabolismo , Polos do Fuso/metabolismo
5.
Mech Dev ; 152: 1-12, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29777776

RESUMO

Contactin2 (Cntn2)/Transient Axonal Glycoprotein 1 (Tag1), a neural cell adhesion molecule, has established roles in neuronal migration and axon fasciculation in chick and mouse. In zebrafish, antisense morpholino-based studies have indicated roles for cntn2 in the migration of facial branchiomotor (FBM) neurons, the guidance of the axons of the nucleus of the medial longitudinal fascicle (nucMLF), and the outgrowth of Rohon-Beard (RB) central axons. To study functions of Cntn2 in later stages of neuronal development, we generated cntn2 mutant zebrafish using CRISPR-Cas9. Using a null mutant allele, we detected genetic interactions between cntn2 and the planar cell polarity gene vangl2, as shown previously with cntn2 morphants, demonstrating a function for cntn2 during FBM neuron migration in a sensitized background of reduced planar cell polarity signaling. In addition, maternal-zygotic (MZ) cntn2 mutant larvae exhibited aberrant touch responses and swimming, suggestive of defects in sensorimotor circuits, consistent with studies in mice. However, the nucMLF axon convergence, FBM neuron migration, and RB outgrowth defects seen in morphants were not seen in the mutants, and we show here that they are likely off-target effects of morpholinos. However, MLF axons exhibited local defasciculation in MZcntn2 mutants, consistent with a role for Cntn2 in axon fasciculation. These data demonstrate distinct roles for zebrafish cntn2 in neuronal migration and axon fasciculation, and in the function of sensorimotor circuits.


Assuntos
Adesão Celular/genética , Contactina 2/genética , Neurogênese/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Axônios/metabolismo , Sistemas CRISPR-Cas , Movimento Celular/genética , Polaridade Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Morfolinos/genética , Morfolinos/metabolismo , Neurônios Motores/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
6.
Front Cell Neurosci ; 11: 107, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28473757

RESUMO

Axon growth and branching, and development of neuronal polarity are critically dependent on proper organization and dynamics of the microtubule (MT) cytoskeleton. MTs must organize with correct polarity for delivery of diverse cargos to appropriate subcellular locations, yet the molecular mechanisms regulating MT polarity remain poorly understood. Moreover, how an actively branching axon reorganizes MTs to direct their plus ends distally at branch points is unknown. We used high-speed, in vivo imaging of polymerizing MT plus ends to characterize MT dynamics in developing sensory axon arbors in zebrafish embryos. We find that axonal MTs are highly dynamic throughout development, and that the peripheral and central axons of sensory neurons show differences in MT behaviors. Furthermore, we show that Calsyntenin-1 (Clstn-1), a kinesin adaptor required for sensory axon branching, also regulates MT polarity in developing axon arbors. In wild type neurons the vast majority of MTs are directed in the correct plus-end-distal orientation from early stages of development. Loss of Clstn-1 causes an increase in MTs polymerizing in the retrograde direction. These misoriented MTs most often are found near growth cones and branch points, suggesting Clstn-1 is particularly important for organizing MT polarity at these locations. Together, our results suggest that Clstn-1, in addition to regulating kinesin-mediated cargo transport, also organizes the underlying MT highway during axon arbor development.

7.
Neural Dev ; 11: 2, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26791407

RESUMO

BACKGROUND: Charcot-Marie-Tooth2b (CMT2b) is an axonal form of a human neurodegenerative disease that preferentially affects sensory neurons. CMT2b is dominantly inherited and is characterized by unusually early onset, presenting in the second or third decade of life. Five missense mutations in the gene encoding Rab7 GTPase have been identified as causative in human CMT2b disease. Although several studies have modeled CMT2b disease in cultured neurons and in Drosophila, the mechanisms by which defective Rab7 leads to disease remain poorly understood. RESULTS: We used zebrafish to investigate the effects of CMT2b-associated Rab7 mutations in a vertebrate model. We generated transgenic animals expressing the CMT2b-associated mutant forms of Rab7 in sensory neurons, and show that these Rab7 variants cause neurodevelopmental defects, including defects in sensory axon growth, branching and pathfinding at early developmental stages. We also find reduced axon growth and branching in neurons expressing a constitutively active form of Rab7, suggesting these defects may be caused by Rab7 gain-of-function. Further, we use high-speed, high-resolution imaging of endosome transport in vivo and find that CMT2b-associated Rab7 variants cause reduced vesicle speeds, suggesting altered transport may underlie axon development defects. CONCLUSIONS: Our data provide new insight into how disease-associated alterations in Rab7 protein disrupt cellular function in vertebrate sensory neurons. Moreover, our findings suggest that defects in axon development may be a previously unrecognized component of CMT2b disease.


Assuntos
Axônios/fisiologia , Doença de Charcot-Marie-Tooth/embriologia , Doença de Charcot-Marie-Tooth/genética , Células Receptoras Sensoriais/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas rab de Ligação ao GTP/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Axônios/patologia , Morte Celular , Doença de Charcot-Marie-Tooth/patologia , Modelos Animais de Doenças , Endossomos/genética , Endossomos/fisiologia , Laminopatias , Dados de Sequência Molecular , Mutação , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Alinhamento de Sequência , Peixe-Zebra , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
8.
J Neurosci ; 34(28): 9235-48, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009257

RESUMO

Precise regulation of axon branching is crucial for neuronal circuit formation, yet the mechanisms that control branch formation are not well understood. Moreover, the highly complex morphology of neurons makes them critically dependent on protein/membrane trafficking and transport systems, although the functions for membrane trafficking in neuronal morphogenesis are largely undefined. Here we identify a kinesin adaptor, Calsyntenin-1 (Clstn-1), as an essential regulator of axon branching and neuronal compartmentalization in vivo. We use morpholino knockdown and a Clstn-1 mutant to show that Clstn-1 is required for formation of peripheral but not central sensory axons, and for peripheral axon branching in zebrafish. We used live imaging of endosomal trafficking in vivo to show that Clstn-1 regulates transport of Rab5-containing endosomes from the cell body to specific locations of developing axons. Our results suggest a model in which Clstn-1 patterns separate axonal compartments and define their ability to branch by directing trafficking of specific endosomes.


Assuntos
Axônios/fisiologia , Axônios/ultraestrutura , Proteínas de Ligação ao Cálcio/metabolismo , Endossomos/fisiologia , Plasticidade Neuronal/fisiologia , Células Receptoras Sensoriais/fisiologia , Células Receptoras Sensoriais/ultraestrutura , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Endossomos/ultraestrutura , Modelos Animais , Neurogênese/fisiologia , Transporte Proteico/fisiologia , Peixe-Zebra
9.
Development ; 141(12): 2506-15, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24917505

RESUMO

The epithelial-to-mesenchymal transition (EMT) is a complex change in cell phenotype that is important for cell migration, morphogenesis and carcinoma metastasis. Loss of epithelial cell adhesion and tight regulation of cadherin adhesion proteins are crucial for EMT. Cells undergoing EMT often display cadherin switching, where they downregulate one cadherin and induce expression of another. However, the functions of the upregulated cadherins and their effects on cell motility are poorly understood. Neural crest cells (NCCs), which undergo EMT during development, lose N-cadherin and upregulate Cadherin 6 (Cdh6) prior to EMT. Cdh6 has been suggested to suppress EMT via cell adhesion, but also to promote EMT by mediating pro-EMT signals. Here, we determine novel roles for Cdh6 in generating cell motility during EMT. We use live imaging of NCC behavior in vivo to show that Cdh6 promotes detachment of apical NCC tails, an important early step of EMT. Furthermore, we show that Cdh6 affects spatiotemporal dynamics of F-actin and active Rho GTPase, and that Cdh6 is required for accumulation of F-actin in apical NCC tails during detachment. Moreover, Cdh6 knockdown alters the subcellular distribution of active Rho, which is known to promote localized actomyosin contraction that is crucial for apical NCC detachment. Together, these data suggest that Cdh6 is an important determinant of where subcellular actomyosin forces are generated during EMT. Our results also identify mechanisms by which an upregulated cadherin can generate cell motility during EMT.


Assuntos
Actinas/metabolismo , Caderinas/fisiologia , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/embriologia , Citoesqueleto de Actina , Actomiosina/metabolismo , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Caderinas/genética , Adesão Celular , Movimento Celular , Transplante de Células , DNA Complementar/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Morfogênese , Crista Neural/citologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Quinases Associadas a rho/metabolismo
10.
Development ; 140(15): 3198-209, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23804498

RESUMO

Epithelial-to-mesenchymal transitions (EMTs) are crucial for morphogenesis and carcinoma metastasis, yet mechanisms controlling the underlying cell behaviors are poorly understood. RhoGTPase signaling has been implicated in EMT; however, previous studies have yielded conflicting results regarding Rho function, and its role in EMT remains poorly understood. Elucidation of precise Rho functions has been challenging because Rho signaling is highly context dependent and its activity is tightly regulated spatiotemporally within the cell. To date, few studies have examined how Rho affects cell motility in intact organisms, and the pattern of Rho activity during motile cell behaviors of EMT has not been determined in any system. Here, we image endogenous active Rho during EMT in vivo, and analyze effects of Rho and Rho-kinase (ROCK) manipulation on cell motility in vivo. We show that Rho is activated in a discrete apical region of premigratory neural crest cells during EMT, and Rho-ROCK signaling is essential for apical detachment and generation of motility within the neuroepithelium, a process that has been poorly understood. Furthermore, we find that Arhgap1 restricts Rho activation to apical areas, and this restriction is necessary for detachment. Our results provide new insight into mechanisms controlling local Rho activation and how it affects dynamic cell behaviors and actomyosin contraction during key steps of EMT in an intact living organism.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Crista Neural/embriologia , Crista Neural/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados , Transição Epitelial-Mesenquimal , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/genética , Técnicas de Silenciamento de Genes , Modelos Neurológicos , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/metabolismo , Crista Neural/citologia , Transdução de Sinais , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/genética , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
11.
Small ; 9(6): 863-9, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23143852

RESUMO

Gold nanoparticles (AuNP) show great potential for diagnostic and therapeutic application in humans. A great number of studies have tested the cytotoxicity of AuNP using cell culture. There is, however, an urgent need to test AuNP in vertebrate animal models that interrogate biodistribution and complex biological traits like organ development, whole body metabolism, and cognitive function. The sheer number of different compounds precludes the use of small rodent model for initial screening. The extended fish embryo test (FET) is used here to bridge the gap between cell culture and small animal models. A study on the toxicity of ultrasmall AuNP in wild type and transgenic zebrafish is presented. FET faithfully reproduce all important findings of a previous study in HeLa cells and add new important information on teratogenicity and hepatotoxicity that could not be gained from studying cultured cells.


Assuntos
Proteínas de Fluorescência Verde/genética , Nanopartículas Metálicas/toxicidade , Animais , Ouro/química , Distribuição Tecidual , Peixe-Zebra
12.
Development ; 139(19): 3590-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22899847

RESUMO

Neurons must develop complex structure to form proper connections in the nervous system. The initiation of axons in defined locations on the cell body and their extension to synaptic targets are critical steps in neuronal morphogenesis, yet the mechanisms controlling axon formation in vivo are poorly understood. The centrosome has been implicated in multiple aspects of neuronal morphogenesis; however, its function in axon development is under debate. Conflicting results from studies of centrosome function in axonogenesis suggest that its role is context dependent and underscore the importance of studying centrosome function as neurons develop in their natural environment. Using live imaging of zebrafish Rohon-Beard (RB) sensory neurons in vivo, we discovered a spatiotemporal relationship between centrosome position and the formation of RB peripheral, but not central, axons. We tested centrosome function by laser ablation and found that centrosome disruption inhibited peripheral axon outgrowth. In addition, we show that centrosome position and motility are regulated by LIM homeodomain transcription factor activity, which is specifically required for the development of RB peripheral axons. Furthermore, we show a correlation between centrosome mislocalization and ectopic axon formation in bashful (laminin alpha 1) mutants. Thus, both intrinsic transcription factor activity and extracellular cues can influence centrosome position and axon formation in vivo. This study presents the first positive association between the centrosome and axon formation in vivo and suggests that the centrosome is important for differential neurite formation in neurons with complex axonal morphologies.


Assuntos
Centrossomo/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas com Domínio LIM/fisiologia , Neuritos/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Axônios/fisiologia , Axônios/ultraestrutura , Centrossomo/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes Dominantes/genética , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Proteínas com Homeodomínio LIM/química , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Proteínas com Homeodomínio LIM/fisiologia , Laminina/genética , Modelos Biológicos , Movimento/fisiologia , Neuritos/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
Neural Dev ; 6: 27, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21619654

RESUMO

BACKGROUND: Development of specific neuronal morphology requires precise control over cell motility processes, including axon formation, outgrowth and branching. Dynamic remodeling of the filamentous actin (F-actin) cytoskeleton is critical for these processes; however, little is known about the mechanisms controlling motile axon behaviors and F-actin dynamics in vivo. Neuronal structure is specified in part by intrinsic transcription factor activity, yet the molecular and cellular steps between transcription and axon behavior are not well understood. Zebrafish Rohon-Beard (RB) sensory neurons have a unique morphology, with central axons that extend in the spinal cord and a peripheral axon that innervates the skin. LIM homeodomain (LIM-HD) transcription factor activity is required for formation of peripheral RB axons. To understand how neuronal morphogenesis is controlled in vivo and how LIM-HD transcription factor activity differentially regulates peripheral versus central axons, we used live imaging of axon behavior and F-actin distribution in vivo. RESULTS: We used an F-actin biosensor containing the actin-binding domain of utrophin to characterize actin rearrangements during specific developmental processes in vivo, including axon initiation, consolidation and branching. We found that peripheral axons initiate from a specific cellular compartment and that F-actin accumulation and protrusive activity precede peripheral axon initiation. Moreover, disruption of LIM-HD transcriptional activity has different effects on the motility of peripheral versus central axons; it inhibits peripheral axon initiation, growth and branching, while increasing the growth rate of central axons. Our imaging revealed that LIM-HD transcription factor activity is not required for F-actin based protrusive activity or F-actin accumulation during peripheral axon initiation, but can affect positioning of F-actin accumulation and axon formation. CONCLUSION: Our ability to image the dynamics of F-actin distribution during neuronal morphogenesis in vivo is unprecedented, and our experiments provide insight into the regulation of cell motility as neurons develop in the intact embryo. We identify specific motile cell behaviors affected by LIM-HD transcription factor activity and reveal how transcription factors differentially control the formation and growth of two axons from the same neuron.


Assuntos
Actinas/metabolismo , Axônios/fisiologia , Movimento Celular/fisiologia , Proteínas de Homeodomínio/metabolismo , Nervos Periféricos/citologia , Células Receptoras Sensoriais/fisiologia , Medula Espinal/citologia , Animais , Animais Geneticamente Modificados , Movimento Celular/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Microscopia Confocal , Pele/inervação , Medula Espinal/embriologia , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
14.
Curr Opin Neurobiol ; 21(1): 17-22, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20970990

RESUMO

Accurate neural crest cell (NCC) migration requires tight control of cell adhesions, cytoskeletal dynamics and cell motility. Cadherins and RhoGTPases are critical molecular players that regulate adhesions and motility during initial delamination of NCCs from the neuroepithelium. Recent studies have revealed multiple functions for these molecules and suggest that a precise balance of their activity is crucial. RhoGTPase appears to regulate both cell adhesions and protrusive forces during NCC delamination. Increasing evidence shows that cadherins are multi-functional proteins with novel, adhesion-independent signaling functions that control NCC motility during both delamination and migration. These functions are often regulated by specific proteolytic cleavage of cadherins. After NCC delamination, planar cell polarity signaling acts via RhoGTPases to control NCC protrusions and migration direction.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Crista Neural/citologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Caderinas/fisiologia , Humanos , Transdução de Sinais , Proteínas rho de Ligação ao GTP/fisiologia
15.
Cell Adh Migr ; 4(4): 586-94, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20671421

RESUMO

Neural crest cells (NCCs) are a remarkable, dynamic group of cells that travel long distances in the embryo to reach their target sites. They are responsible for the formation of craniofacial bones and cartilage, neurons and glia in the peripheral nervous system, and pigment cells. Live imaging of NCCs as they traverse the embryo has been critical to increasing our knowledge of their biology. NCCs exhibit multiple behaviors and communicate with each other and their environment along each step of their journey. Imaging combined with molecular manipulations has led to insights into the mechanisms controlling these behaviors. In this review, we highlight studies that have used live imaging to provide novel insight into NCC migration and discuss how continued use of such techniques can advance our understanding of NCC biology.


Assuntos
Movimento Celular , Rastreamento de Células , Crista Neural/citologia , Animais , Efrinas/metabolismo , Transição Epitelial-Mesenquimal , Tubo Neural/embriologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
16.
J Neurosci ; 29(42): 13190-201, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19846707

RESUMO

Multiple molecular cues guide neuronal axons to their targets during development. Previous studies in vitro have shown that mechanical stimulation also can affect axon growth; however, whether mechanical force contributes to axon guidance in vivo is unknown. We investigated the role of muscle contractions in the guidance of zebrafish peripheral Rohon-Beard (RB) sensory axons in vivo. We analyzed several mutants that affect muscle contraction through different molecular pathways, including a new mutant allele of the titin a (pik) gene, mutants that affect the hedgehog signaling pathway, and a nicotinic acetylcholine receptor mutant. We found RB axon defects in these mutants, the severity of which appeared to correlate with the extent of muscle contraction loss. These axons extend between the muscle and skin and normally have ventral trajectories and repel each other on contact. RB peripheral axons in muscle mutants extend longitudinally instead of ventrally, and the axons fail to repel one another on contact. In addition, we showed that limiting muscle movements by embedding embryos in agarose caused similar defects in peripheral RB axon guidance. This work suggests that the mechanical forces generated by muscle contractions are necessary for proper sensory axon pathfinding in vivo.


Assuntos
Axônios/fisiologia , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Nervos Periféricos/citologia , Células Receptoras Sensoriais/citologia , Animais , Animais Geneticamente Modificados , Axônios/efeitos dos fármacos , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Condroitina ABC Liase/farmacologia , Conectina , Embrião não Mamífero/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/metabolismo , Contração Muscular/efeitos dos fármacos , Proteínas Musculares/genética , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/embriologia , Mutação/genética , Fármacos Neuromusculares não Despolarizantes/farmacologia , Proteínas Quinases/genética , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Tempo , Tubocurarina/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
17.
Dev Biol ; 325(2): 363-73, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19013446

RESUMO

Interactions between a neuron and its environment play a major role in neuronal migration. We show here that the cell adhesion molecule Transient Axonal Glycoprotein (Tag1) is necessary for the migration of the facial branchiomotor neurons (FBMNs) in the zebrafish hindbrain. In tag1 morphant embryos, FBMN migration is specifically blocked, with no effect on organization or patterning of other hindbrain neurons. Furthermore, using suboptimal morpholino doses and genetic mutants, we found that tag1, lamininalpha1 (lama1) and stbm, which encodes a transmembrane protein Vangl2, exhibit pairwise genetic interactions for FBMN migration. Using time-lapse analyses, we found that FBMNs are affected similarly in all three single morphant embryos, with an inability to extend protrusions in a specific direction, and resulting in the failure of caudal migration. These data suggest that tag1, lama1 and vangl2 participate in a common mechanism that integrates signaling between the FBMN and its environment to regulate migration.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Laminina/fisiologia , Proteínas de Membrana/fisiologia , Neurônios Motores/fisiologia , Rombencéfalo/embriologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Adesão Celular , Movimento Celular , Contactina 2 , Rombencéfalo/fisiologia , Transdução de Sinais , Peixe-Zebra/fisiologia
18.
Dev Biol ; 324(2): 236-44, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18926812

RESUMO

The induction and migration of neural crest cells (NCCs) are essential to the development of craniofacial structures and the peripheral nervous system. A critical step in the development of NCCs is the epithelial to mesenchymal transition (EMT) that they undergo in order to initiate migration. Several transcription factors are important for the NCC EMT. However, less is known about the effectors regulating changes in cell adhesion, the cytoskeleton, and cell motility associated with the EMT or about specific changes in the behavior of cells undergoing EMT in vivo. We used time-lapse imaging of NCCs in the zebrafish hindbrain to show that NCCs undergo a stereotypical series of behaviors during EMT. We find that loss of cell adhesion and membrane blebbing precede filopodial extension and the onset of migration. Live imaging of actin dynamics shows that actin localizes differently in blebs and filopodia. Moreover, we find that disruption of myosin II or Rho-kinase (ROCK) activity inhibits NCC blebbing and causes reduced NCC EMT. These data reveal roles for myosin II and ROCK in NCC EMT in vivo, and provide a detailed characterization of NCC behavior during EMT that will form a basis for further mechanistic studies.


Assuntos
Diferenciação Celular , Miosina Tipo II/metabolismo , Crista Neural/embriologia , Peixe-Zebra/embriologia , Quinases Associadas a rho/metabolismo , Actinas/metabolismo , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/metabolismo , Adesão Celular , Membrana Celular/metabolismo , Movimento Celular , Citocinese , Epitélio/embriologia , Mesoderma/embriologia , Crista Neural/ultraestrutura , Pseudópodes/fisiologia , Rombencéfalo/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
19.
Dev Dyn ; 237(6): 1645-52, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18498099

RESUMO

In the anterior vertebrate head, a population of neural crest cells (NCCs) migrates to the periocular mesenchyme and makes critical contributions to the developing eye and orbit. Improper migration and differentiation of these NCCs have been implicated in human diseases such as congenital glaucoma and anterior segment dysgenesis syndromes. The mechanisms by which these cells migrate to their target tissues within and around the eye are not well understood. We present a fate map of zebrafish diencephalic and mesencephalic NCC contributions to the eye and orbit. The fate map closely resembles that in chick and mice, demonstrating evolutionary conservation. To gain insight into the mechanisms of anterior NCC guidance, we used the eyeless mutant chokh/rx3. We show that, in chokh mutants, dorsal anterior NCC migration is severely disorganized. Time-lapse analysis shows that NCCs have significantly reduced migration rates and directionality in chokh mutants.


Assuntos
Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/embriologia , Animais , Cartilagem/metabolismo , Movimento Celular , Evolução Molecular , Perfilação da Expressão Gênica , Hibridização In Situ , Mesoderma , Modelos Biológicos , Mutação , Crista Neural/citologia , Fatores de Tempo , Distribuição Tecidual , Peixe-Zebra
20.
Neural Dev ; 3: 6, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18289389

RESUMO

BACKGROUND: How axon guidance signals regulate growth cone behavior and guidance decisions in the complex in vivo environment of the central nervous system is not well understood. We have taken advantage of the unique features of the zebrafish embryo to visualize dynamic growth cone behaviors and analyze guidance mechanisms of axons emerging from a central brain nucleus in vivo. RESULTS: We investigated axons of the nucleus of the medial longitudinal fascicle (nucMLF), which are the first axons to extend in the zebrafish midbrain. Using in vivo time-lapse imaging, we show that both positive axon-axon interactions and guidance by surrounding tissue control initial nucMLF axon guidance. We further show that two guidance molecules, transient axonal glycoprotein-1 (TAG-1) and laminin-alpha1, are essential for the initial directional extension of nucMLF axons and their subsequent convergence into a tight fascicle. Fixed tissue analysis shows that TAG-1 knockdown causes errors in nucMLF axon pathfinding similar to those seen in a laminin-alpha1 mutant. However, in vivo time-lapse imaging reveals that while some defects in dynamic growth cone behavior are similar, there are also defects unique to the loss of each gene. Loss of either TAG-1 or laminin-alpha1 causes nucMLF axons to extend into surrounding tissue in incorrect directions and reduces axonal growth rate, resulting in stunted nucMLF axons that fail to extend beyond the hindbrain. However, defects in axon-axon interactions were found only after TAG-1 knockdown, while defects in initial nucMLF axon polarity and excessive branching of nucMLF axons occurred only in laminin-alpha1 mutants. CONCLUSION: These results demonstrate how two guidance cues, TAG-1 and laminin-alpha1, influence the behavior of growth cones during axon pathfinding in vivo. Our data suggest that TAG-1 functions to allow growth cones to sense environmental cues and mediates positive axon-axon interactions. Laminin-alpha1 does not regulate axon-axon interactions, but does influence neuronal polarity and directional guidance.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Cones de Crescimento/fisiologia , Laminina/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Axônios/fisiologia , Contactina 2 , Regulação da Expressão Gênica no Desenvolvimento , Mesencéfalo/embriologia , Mesencéfalo/fisiologia , Mutagênese , Vias Neurais/citologia , Vias Neurais/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA