Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 615(7952): 402-403, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36859662
2.
Hum Reprod ; 38(4): 655-670, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36807972

RESUMO

STUDY QUESTION: Is the vertebrate protein Dead end (DND1) a causative factor for human infertility and can novel in vivo assays in zebrafish help in evaluating this? SUMMARY ANSWER: Combining patient genetic data with functional in vivo assays in zebrafish reveals a possible role for DND1 in human male fertility. WHAT IS KNOWN ALREADY: About 7% of the male population is affected by infertility but linking specific gene variants to the disease is challenging. The function of the DND1 protein was shown to be critical for germ cell development in several model organisms but a reliable and cost-effective method for evaluating the activity of the protein in the context of human male infertility is still missing. STUDY DESIGN, SIZE, DURATION: Exome data from 1305 men included in the Male Reproductive Genomics cohort were examined in this study. A total of 1114 of the patients showed severely impaired spermatogenesis but were otherwise healthy. Eighty-five men with intact spermatogenesis were included in the study as controls. PARTICIPANTS/MATERIALS, SETTING, METHODS: We screened the human exome data for rare, stop-gain, frameshift, splice site, as well as missense variants in DND1. The results were validated by Sanger sequencing. Immunohistochemical techniques and, when possible, segregation analyses were performed for patients with identified DND1 variants. The amino acid exchange in the human variant was mimicked at the corresponding site of the zebrafish protein. Using different aspects of germline development in live zebrafish embryos as biological assays, we examined the activity level of these DND1 protein variants. MAIN RESULTS AND THE ROLE OF CHANCE: In human exome sequencing data, we identified four heterozygous variants in DND1 (three missense and one frameshift variant) in five unrelated patients. The function of all of the variants was examined in the zebrafish and one of those was studied in more depth in this model. We demonstrate the use of zebrafish assays as a rapid and effective biological readout for evaluating the possible impact of multiple gene variants on male fertility. This in vivo approach allowed us to assess the direct impact of the variants on germ cell function in the context of the native germline. Focusing on the DND1 gene, we find that zebrafish germ cells, expressing orthologs of DND1 variants identified in infertile men, failed to arrive correctly at the position where the gonad develops and exhibited defects in cell fate maintenance. Importantly, our analysis facilitated the evaluation of single nucleotide variants, whose impact on protein function is difficult to predict, and allowed us to distinguish variants that do not affect the protein's activity from those that strongly reduce it and could thus potentially be the primary cause for the pathological condition. These aberrations in germline development resemble the testicular phenotype of azoospermic patients. LIMITATIONS, REASONS FOR CAUTION: The pipeline we present requires access to zebrafish embryos and to basic imaging equipment. The notion that the activity of the protein in the zebrafish-based assays is relevant for the human homolog is well supported by previous knowledge. Nevertheless, the human protein may differ in some respects from its homologue in zebrafish. Thus, the assay should be considered only one of the parameters used in defining DND1 variants as causative or non-causative for infertility. WIDER IMPLICATIONS OF THE FINDINGS: Using DND1 as an example, we have shown that the approach described in this study, relying on bridging between clinical findings and fundamental cell biology, can help to establish links between novel human disease candidate genes and fertility. In particular, the power of the approach we developed is manifested by the fact that it allows the identification of DND1 variants that arose de novo. The strategy presented here can be applied to different genes in other disease contexts. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by the German Research Foundation, Clinical Research Unit, CRU326 'Male Germ Cells'. There are no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Infertilidade Masculina , Peixe-Zebra , Animais , Humanos , Masculino , Peixe-Zebra/genética , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Testículo/patologia , Fertilidade , Fenótipo , Proteínas de Neoplasias/genética
3.
Dev Cell ; 57(16): 2026-2040.e5, 2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-35914525

RESUMO

Cell ablation is a key method in the research fields of developmental biology, tissue regeneration, and tissue homeostasis. Eliminating specific cell populations allows for characterizing interactions that control cell differentiation, death, behavior, and spatial organization of cells. Current methodologies for inducing cell death suffer from relatively slow kinetics, making them unsuitable for analyzing rapid events and following primary and immediate consequences of the ablation. To address this, we developed a cell-ablation system that is based on bacterial toxin/anti-toxin proteins and enables rapid and cell-autonomous elimination of specific cell types and organs in zebrafish embryos. A unique feature of this system is that it uses an anti-toxin, which allows for controlling the degree and timing of ablation and the resulting phenotypes. The transgenic zebrafish generated in this work represent a highly efficient tool for cell ablation, and this approach is applicable to other model organisms as demonstrated here for Drosophila.


Assuntos
Drosophila , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Morte Celular , Diferenciação Celular , Peixe-Zebra/genética
4.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33574063

RESUMO

To study the mechanisms controlling front-rear polarity in migrating cells, we used zebrafish primordial germ cells (PGCs) as an in vivo model. We find that polarity of bleb-driven migrating cells can be initiated at the cell front, as manifested by actin accumulation at the future leading edge and myosin-dependent retrograde actin flow toward the other side of the cell. In such cases, the definition of the cell front, from which bleb-inhibiting proteins such as Ezrin are depleted, precedes the establishment of the cell rear, where those proteins accumulate. Conversely, following cell division, the accumulation of Ezrin at the cleavage plane is the first sign for cell polarity and this aspect of the cell becomes the cell back. Together, the antagonistic interactions between the cell front and back lead to a robust polarization of the cell. Furthermore, we show that chemokine signaling can bias the establishment of the front-rear axis of the cell, thereby guiding the migrating cells toward sites of higher levels of the attractant. We compare these results to a theoretical model according to which a critical value of actin treadmilling flow can initiate a positive feedback loop that leads to the generation of the front-rear axis and to stable cell polarization. Together, our in vivo findings and the mathematical model, provide an explanation for the observed nonoriented migration of primordial germ cells in the absence of the guidance cue, as well as for the directed migration toward the region where the gonad develops.


Assuntos
Actinas/metabolismo , Movimento Celular , Polaridade Celular , Quimiocinas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Transporte Proteico , Peixe-Zebra
5.
Dev Cell ; 43(6): 704-715.e5, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29257950

RESUMO

Maintaining cell fate relies on robust mechanisms that prevent the differentiation of specified cells into other cell types. This is especially critical during embryogenesis, when extensive cell proliferation, patterning, and migration events take place. Here we show that vertebrate primordial germ cells (PGCs) are protected from reprogramming into other cell types by the RNA-binding protein Dead end (Dnd). PGCs knocked down for Dnd lose their characteristic morphology and adopt various somatic cell fates. Concomitantly, they gain a gene expression profile reflecting differentiation into cells of different germ layers, in a process that we could direct by expression of specific cell-fate determinants. Importantly, we visualized these events within live zebrafish embryos, which provide temporal information regarding cell reprogramming. Our results shed light on the mechanisms controlling germ cell fate maintenance and are relevant for the formation of teratoma, a tumor class composed of cells from more than one germ layer.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Diferenciação Celular/genética , Movimento Celular , Técnicas de Reprogramação Celular/métodos , Endoderma/fisiologia , Células Germinativas/metabolismo , Células Germinativas/fisiologia , Hibridização In Situ , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
6.
Dev Cell ; 43(5): 577-587.e5, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29173819

RESUMO

Cell migration is essential for morphogenesis, organ formation, and homeostasis, with relevance for clinical conditions. The migration of primordial germ cells (PGCs) is a useful model for studying this process in the context of the developing embryo. Zebrafish PGC migration depends on the formation of cellular protrusions in form of blebs, a type of protrusion found in various cell types. Here we report on the mechanisms allowing the inflation of the membrane during bleb formation. We show that the rapid expansion of the protrusion depends on membrane invaginations that are localized preferentially at the cell front. The formation of these invaginations requires the function of Cdc42, and their unfolding allows bleb inflation and dynamic cell-shape changes performed by migrating cells. Inhibiting the formation and release of the invaginations strongly interfered with bleb formation, cell motility, and the ability of the cells to reach their target.


Assuntos
Membrana Celular/metabolismo , Movimento Celular/fisiologia , Forma Celular/fisiologia , Células Germinativas/citologia , Peixe-Zebra , Actinas/metabolismo , Animais , Estruturas da Membrana Celular/metabolismo , Extensões da Superfície Celular/metabolismo , Células Germinativas/metabolismo , Peixe-Zebra/metabolismo
7.
Nat Cell Biol ; 18(4): 366-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27027487

RESUMO

A powerful combination of cell labelling, genetic tools and rapid imaging techniques in vivo has now led to a high-resolution description of lumen formation during angiogenesis in zebrafish. The study reveals a haemodynamic-force-driven and myosin-II-dependent cellular mechanism (termed inverse membrane blebbing) as the basis for lumen expansion in unicellular and multicellular angiogenic sprouts.


Assuntos
Vasos Sanguíneos/embriologia , Morfogênese , Neovascularização Fisiológica , Animais , Feminino , Masculino
8.
Curr Biol ; 25(8): 1096-103, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25843033

RESUMO

Directional cell migration requires cell polarization with respect to the distribution of the guidance cue. Cell polarization often includes asymmetric distribution of response components as well as elements of the motility machinery. Importantly, the function and regulation of most of these molecules are known to be pH dependent. Intracellular pH gradients were shown to occur in certain cells migrating in vitro, but the functional relevance of such gradients for cell migration and for the response to directional cues, particularly in the intact organism, is currently unknown. In this study, we find that primordial germ cells migrating in the context of the developing embryo respond to the graded distribution of the chemokine Cxcl12 by establishing elevated intracellular pH at the cell front. We provide insight into the mechanisms by which a polar pH distribution contributes to efficient cell migration. Specifically, we show that Carbonic Anhydrase 15b, an enzyme controlling the pH in many cell types, including metastatic cancer cells, is expressed in migrating germ cells and is crucial for establishing and maintaining an asymmetric pH distribution within them. Reducing the level of the protein and thereby erasing the pH elevation at the cell front resulted in abnormal cell migration and impaired arrival at the target. The basis for the disrupted migration is found in the stringent requirement for pH conditions in the cell for regulating contractility, for the polarization of Rac1 activity, and hence for the formation of actin-rich structures at the leading edge of the migrating cells.


Assuntos
Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Quimiocinas/metabolismo , Células Germinativas/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Líquido Intracelular/química , Actinas/metabolismo , Animais , Quimiocina CXCL12/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Células Germinativas/metabolismo , Líquido Intracelular/metabolismo , Peixe-Zebra
9.
Elife ; 42015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25875301

RESUMO

Cell migration and polarization is controlled by signals in the environment. Migrating cells typically form filopodia that extend from the cell surface, but the precise function of these structures in cell polarization and guided migration is poorly understood. Using the in vivo model of zebrafish primordial germ cells for studying chemokine-directed single cell migration, we show that filopodia distribution and their dynamics are dictated by the gradient of the chemokine Cxcl12a. By specifically interfering with filopodia formation, we demonstrate for the first time that these protrusions play an important role in cell polarization by Cxcl12a, as manifested by elevation of intracellular pH and Rac1 activity at the cell front. The establishment of this polarity is at the basis of effective cell migration towards the target. Together, we show that filopodia allow the interpretation of the chemotactic gradient in vivo by directing single-cell polarization in response to the guidance cue.


Assuntos
Movimento Celular , Polaridade Celular , Quimiocina CXCL12/metabolismo , Células Germinativas/citologia , Espaço Intracelular/metabolismo , Pseudópodes/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Embrião não Mamífero/metabolismo , Endocitose/efeitos dos fármacos , Células Germinativas/metabolismo , Modelos Biológicos , Receptores CXCR4/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
10.
Development ; 141(16): 3188-96, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063456

RESUMO

Collective migration of cells in the zebrafish posterior lateral line primordium (PLLp) along a path defined by Cxcl12a expression depends on Cxcr4b receptors in leading cells and on Cxcr7b in trailing cells. Cxcr7b-mediated degradation of Cxcl12a by trailing cells generates a local gradient of Cxcl12a that guides PLLp migration. Agent-based computer models were built to explore how a polarized response to Cxcl12a, mediated by Cxcr4b in leading cells and prevented by Cxcr7b in trailing cells, determines unidirectional migration of the PLLp. These chemokine signaling-based models effectively recapitulate many behaviors of the PLLp and provide potential explanations for the characteristic behaviors that emerge when the PLLp is severed by laser to generate leading and trailing fragments. As predicted by our models, the bilateral stretching of the leading fragment is lost when chemokine signaling is blocked in the PLLp. However, movement of the trailing fragment toward the leading cells, which was also thought to be chemokine dependent, persists. This suggested that a chemokine-independent mechanism, not accounted for in our models, is responsible for this behavior. Further investigation of trailing cell behavior shows that their movement toward leading cells depends on FGF signaling and it can be re-oriented by exogenous FGF sources. Together, our observations reveal the simple yet elegant manner in which leading and trailing cells coordinate migration; while leading cells steer PLLp migration by following chemokine cues, cells further back play follow-the-leader as they migrate toward FGFs produced by leading cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sistema da Linha Lateral/embriologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Comunicação Celular , Movimento Celular , Quimiocina CXCL12/fisiologia , Quimiocinas/metabolismo , Simulação por Computador , Fatores de Crescimento de Fibroblastos/metabolismo , Receptores CXCR/fisiologia , Receptores CXCR4/fisiologia , Transdução de Sinais , Proteínas de Peixe-Zebra/fisiologia
11.
Bioessays ; 36(8): 741-5, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24924601

RESUMO

The identification of molecules controlling embryonic patterning and their functional analysis has revolutionized the fields of Developmental and Cell Biology. The use of new sequence information and modern bioinformatics tools has enriched the list of proteins that could potentially play a role in regulating cell behavior and function during early development. The recent application of efficient methods for gene knockout in zebrafish has accelerated the functional analysis of many proteins, some of which have been overlooked due to their small size. Two recent publications report on the identification of one such protein and its role in zebrafish embryogenesis. The protein, currently designated Apela, was shown to act as a secreted protein whose absence adversely affected various early developmental processes. Additional signaling proteins that have been identified in one of the studies are likely to open the way to unraveling hitherto unknown developmental pathways and have the potential to provide a more comprehensive understanding of known developmental processes.


Assuntos
Transdução de Sinais , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/metabolismo , Animais , Receptores de Apelina , Gástrula/metabolismo , Gastrulação , Coração/embriologia , Humanos , Organogênese , Receptores Acoplados a Proteínas G/fisiologia , Peixe-Zebra/embriologia
12.
Nat Cell Biol ; 12(1): 47-53; sup pp 1-11, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20010816

RESUMO

Cell migration is central to embryonic development, homeostasis and disease, processes in which cells move as part of a group or individually. Whereas the mechanisms controlling single-cell migration in vitro are relatively well understood, less is known about the mechanisms promoting the motility of individual cells in vivo. In particular, it is not clear how cells that form blebs in their migration use those protrusions to bring about movement in the context of the three-dimensional cellular environment. Here we show that the motility of chemokine-guided germ cells within the zebrafish embryo requires the function of the small Rho GTPases Rac1 and RhoA, as well as E-cadherin-mediated cell-cell adhesion. Using fluorescence resonance energy transfer we demonstrate that Rac1 and RhoA are activated in the cell front. At this location, Rac1 is responsible for the formation of actin-rich structures, and RhoA promotes retrograde actin flow. We propose that these actin-rich structures undergoing retrograde flow are essential for the generation of E-cadherin-mediated traction forces between the germ cells and the surrounding tissue and are therefore crucial for cell motility in vivo.


Assuntos
Caderinas/metabolismo , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células Germinativas/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Caderinas/genética , Polaridade Celular , Células Cultivadas , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Transferência Ressonante de Energia de Fluorescência , Peixe-Zebra
13.
Cell Host Microbe ; 6(2): 174-86, 2009 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-19683683

RESUMO

HIV-1 Nef is a key factor in AIDS pathogenesis. Here, we report that Nef potently inhibits motility of fibroblasts and chemotaxis of HIV-1-infected primary human T lymphocytes toward the chemokines SDF-1alpha, CCL-19, and CCL-21 ex vivo. Furthermore, Nef inhibits guided motility of zebrafish primordial germ cells toward endogenous SDF-1a in vivo. These migration defects result from Nef-mediated inhibition of the actin remodeling normally triggered by migratory stimuli. Nef strongly induces phosphorylation of cofilin, inactivating this evolutionarily conserved actin-depolymerizing factor that promotes cell motility when unphosphorylated. Nef-dependent cofilin deregulation requires association of Nef with the cellular kinase Pak2. Disruption of Nef-Pak2 association restores the cofilin phosphorylation levels and actin remodeling that facilitate cell motility. We conclude that HIV-1 Nef alters Pak2 function, which directly or indirectly inactivates cofilin, thereby restricting migration of infected T lymphocytes as part of a strategy to optimize immune evasion and HIV-1 replication.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Movimento Celular , HIV-1/patogenicidade , Fatores de Virulência/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Linhagem Celular , Células Cultivadas , Fibroblastos/virologia , Humanos , Linfócitos T/virologia , Quinases Ativadas por p21/metabolismo
14.
Cell ; 132(3): 463-73, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18267076

RESUMO

Primordial germ cell (PGC) migration in zebrafish is directed by the chemokine SDF-1a that activates its receptor CXCR4b. Little is known about the molecular mechanisms controlling the distribution of this chemoattractant in vivo. We demonstrate that the activity of a second SDF-1/CXCL12 receptor, CXCR7, is crucial for proper migration of PGCs toward their targets. We show that CXCR7 functions primarily in the somatic environment rather than within the migrating cells. In CXCR7 knocked-down embryos, the PGCs exhibit a phenotype that signifies defects in SDF-1a gradient formation as the cells fail to polarize effectively and to migrate toward their targets. Indeed, somatic cells expressing CXCR7 show enhanced internalization of the chemokine suggesting that CXCR7 acts as a sink for SDF-1a, thus allowing the dynamic changes in the transcription of sdf-1a to be mirrored by similar dynamics at the protein level.


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Células Germinativas/citologia , Receptores CXCR/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Polaridade Celular , Embrião não Mamífero/citologia , Regulação da Expressão Gênica no Desenvolvimento , Receptores CXCR/genética , Proteínas de Peixe-Zebra/genética
15.
Curr Biol ; 17(13): 1164-72, 2007 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-17600713

RESUMO

Activation of the chemokine receptor CXCR4 by SDF1 controls a variety of biological processes in development, immune response, and disease [1-5]. The carboxyl-terminal region of CXCR4 is subject to phosphorylation that allows binding of regulatory proteins [5]; this results in downregulation of CXCR4 signaling and receptor internalization [6]. Notably, truncations of this part of CXCR4 have been implicated in WHIM syndrome, a dominantly inherited immunodeficiency disorder [7, 8]. Despite its importance in receptor signaling and the clinical relevance of its regulation, the precise function of regulating signaling level and internalization in controlling cell behavior is not known. Whereas a number of in vitro studies suggested that the carboxyl terminus of CXCR4 positively regulates chemotaxis (e.g., [9]), others reached the opposite conclusion [8, 10, 11]. These conflicting results highlight the importance of investigating this process under physiological conditions in the live animal. In this study, we demonstrate the significance of internalization and of controlling receptor signaling level for SDF-1-guided migration. We found that whereas internalization and the control over signaling intensity are dispensable for cell motility and directional sensing, they are essential for fine-tuning of migration in vivo, allowing precise arrival of zebrafish PGCs at their target, the region where the gonad develops.


Assuntos
Movimento Celular/fisiologia , Quimiocina CXCL12/metabolismo , Desenvolvimento Embrionário/fisiologia , Células Germinativas/fisiologia , Receptores CXCR4/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Células Germinativas/metabolismo , Gônadas/embriologia , Estrutura Terciária de Proteína , Serina/metabolismo , Peixe-Zebra
16.
Dev Cell ; 11(5): 613-27, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17084355

RESUMO

The molecular and cellular mechanisms governing cell motility and directed migration in response to the chemokine SDF-1 are largely unknown. Here, we demonstrate that zebrafish primordial germ cells whose migration is guided by SDF-1 generate bleb-like protrusions that are powered by cytoplasmic flow. Protrusions are formed at sites of higher levels of free calcium where activation of myosin contraction occurs. Separation of the acto-myosin cortex from the plasma membrane at these sites is followed by a flow of cytoplasm into the forming bleb. We propose that polarized activation of the receptor CXCR4 leads to a rise in free calcium that in turn activates myosin contraction in the part of the cell responding to higher levels of the ligand SDF-1. The biased formation of new protrusions in a particular region of the cell in response to SDF-1 defines the leading edge and the direction of cell migration.


Assuntos
Quimiotaxia , Citoplasma/fisiologia , Células Germinativas/fisiologia , Miosinas/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Actinas/fisiologia , Animais , Membrana Celular/fisiologia , Polaridade Celular , Quimiocina CXCL12 , Quimiocinas CXC/fisiologia , Citoesqueleto/fisiologia , Pseudópodes/fisiologia , Receptores CXCR4/fisiologia , Peixe-Zebra/embriologia
17.
Curr Opin Genet Dev ; 16(4): 355-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16806897

RESUMO

The migration of zebrafish primordial germ cell towards the region where the gonad develops is guided by the chemokine SDF-1a. Recent studies show that soon after their specification, the cells undergo a series of morphological alterations before they become motile and are able to respond to attractive cues. As migratory cells, primordial germ cells move towards their target while correcting their path upon exiting a cyclic phase in which morphological cell polarity is lost. In the following stages, the cells gather at specific locations and move as cell clusters towards their final target. In all of these stages, zebrafish germ cells respond as individual cells to alterations in the shape of the sdf-1a expression domain, by directed migration towards their target - the position where the gonad develops.


Assuntos
Movimento Celular , Quimiocinas CXC/fisiologia , Células Germinativas/fisiologia , Peixe-Zebra/embriologia , Animais , Quimiocina CXCL12 , Quimiocinas CXC/genética
18.
J Cell Sci ; 118(Pt 17): 4027-38, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16129886

RESUMO

The migration of zebrafish primordial germ cells (PGCs) is directed by SDF-1a and serves as a model for long-range chemokine-guided cell migration. Whereas the development and migration of zebrafish PGCs have been studied in great detail starting at mid-gastrulation stages when the cells exhibit guided active migration [7-8 hours post fertilization (hpf)], earlier stages have not yet been examined. Here we show that the PGCs acquire competence to respond to the chemokine following discrete maturation steps. Using the promoter of the novel gene askopos and RNA elements of nanos1 to drive GFP expression in PGCs, we found that immediately after their specification (about 3 hpf) PGCs exhibit simple cell shape. This stage is followed by a phase at which the cells assume complex morphology yet they neither change their position nor do they respond to SDF-1a. During the third phase, a transition into a ;migratory stage' occurs as PGCs become responsive to directional cues provided by somatic cells secreting the chemokine SDF-1a. This transition depends on zygotic transcription and on the function of the RNA-binding protein Dead end and is correlated with down regulation of the cell adhesion molecule E-cadherin. These distinctive morphological and molecular alterations could represent a general occurrence in similar processes critical for development and disease.


Assuntos
Movimento Celular/fisiologia , Quimiocinas CXC/metabolismo , Células Germinativas/citologia , Células Germinativas/fisiologia , Proteínas de Xenopus/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Caderinas/genética , Caderinas/metabolismo , Caspases/metabolismo , Forma Celular , Quimiocina CXCL12 , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mutagênese Sítio-Dirigida , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transcrição Gênica , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
19.
Dev Cell ; 6(4): 589-96, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15068797

RESUMO

Zebrafish primordial germ cells (PGCs) are guided toward their targets by the chemokine SDF-1a. PGCs were followed during three phases of their migration: when migrating as individual cells, while remaining in a clustered configuration, and when moving as a cell cluster within the embryo. We found that individually migrating PGCs alternate between migratory and pausing modes. Pausing intervals are characterized by loss of cell polarity and correlate with subsequent changes in the direction of migration. These properties constitute an intrinsic behavior of PGCs, enabling erasure of prior polarity and re-sampling of the environment. Following migration arrest at a site of high SDF-1a levels, PGCs resume migration as a cluster. The seemingly coordinated cluster migration is a result of single-cell movement in response to local variations in SDF-1a distribution. Together, these behavioral modes allow the cells to arrive at specific destinations with high fidelity and remain at their target site.


Assuntos
Movimento Celular/genética , Embrião não Mamífero/embriologia , Genitália/embriologia , Células Germinativas/fisiologia , Organogênese/genética , Peixe-Zebra/embriologia , Animais , Comunicação Celular/genética , Diferenciação Celular/genética , Polaridade Celular/genética , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Quimiotaxia/genética , Sinais (Psicologia) , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Genitália/citologia , Genitália/fisiologia , Células Germinativas/citologia
20.
Dev Biol ; 269(2): 567-79, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15110720

RESUMO

During early vertebrate development, ANF homeobox genes are expressed in the prospective forebrain. Their regulation is essential for correct morphogenesis and function of the prosencephalon. We identified a 1-kb fragment upstream of the chicken GANF gene sufficient to drive lacZ expression in the endogenous expression domain. Concordant with the high conservation of this sequence in five investigated species, this element is also active in the corresponding expression domain of the zebrafish orthologue. In vivo analysis of two in vitro-identified Otx2 binding sites in this conserved sequence revealed their necessity for activation of the chicken ANF promoter. In addition, we identified a Pax6-binding site close to the transcriptional start site that is occupied in vivo by Pax6 protein. Pax6 and GANF exhibit mutually exclusive expression domains in the anterior embryonic region. Overexpression of Pax6 in chick embryos inhibited the endogenous GANF expression, and in Pax6(-/-) mice the expression domain of the murine ANF orthologue Hesx1 was expanded and sustained, indicating inhibitory effects of Pax6 on GANF. However, a mutation of the Pax6 site did not abolish reporter activity from an electroporated vector. We conclude that Otx2 and Pax6 are key molecules involved in conserved mechanisms of ANF gene regulation.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Prosencéfalo/embriologia , Transativadores/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Embrião de Galinha , Sequência Conservada , Proteínas do Olho , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/química , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição Otx , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Regiões Promotoras Genéticas , Prosencéfalo/metabolismo , Proteínas Repressoras , Fatores de Transcrição HES-1 , Peixe-Zebra , Proteínas de Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA