Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
PLoS Genet ; 18(7): e1010329, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35877760

RESUMO

Glioblastoma (GB) is the most aggressive, lethal and frequent primary brain tumor. It originates from glial cells and is characterized by rapid expansion through infiltration. GB cells interact with the microenvironment and healthy surrounding tissues, mostly neurons and vessels. GB cells project tumor microtubes (TMs) contact with neurons, and exchange signaling molecules related to Wingless/WNT, JNK, Insulin or Neuroligin-3 pathways. This cell to cell communication promotes GB expansion and neurodegeneration. Moreover, healthy neurons form glutamatergic functional synapses with GB cells which facilitate GB expansion and premature death in mouse GB xerograph models. Targeting signaling and synaptic components of GB progression may become a suitable strategy against glioblastoma. In a Drosophila GB model, we have determined the post-synaptic nature of GB cells with respect to neurons, and the contribution of post-synaptic genes expressed in GB cells to tumor progression. In addition, we document the presence of intratumoral synapses between GB cells, and the functional contribution of pre-synaptic genes to GB calcium dependent activity and expansion. Finally, we explore the relevance of synaptic genes in GB cells to the lifespan reduction caused by GB advance. Our results indicate that both presynaptic and postsynaptic proteins play a role in GB progression and lethality.


Assuntos
Glioblastoma , Animais , Drosophila/genética , Glioblastoma/metabolismo , Camundongos , Neurônios/metabolismo , Transdução de Sinais/genética , Sinapses/metabolismo , Microambiente Tumoral
2.
Dis Model Mech ; 14(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34061177

RESUMO

The mammalian central nervous system (CNS) exhibits limited regenerative capacity and the mechanisms that mediate its regeneration are not fully understood. Here, we present a novel experimental design to damage the CNS by using a contusion injury paradigm. The design of this protocol allows the study of long-term and short-term cellular responses, including those of the CNS and the immune system, and of any implications regarding functional recovery. We demonstrate for the first time that adult Drosophilamelanogaster glial cells undergo spontaneous functional recovery following crush injury. This crush injury leads to an intermediate level of functional recovery after damage, which is ideal to screen for genes that facilitate or prevent the regeneration process. Here, we validate this model and analyse the immune responses of glial cells as a central regulator of functional regeneration. Additionally, we demonstrate that glial cells and macrophages contribute to functional regeneration through mechanisms involving the Jun N-terminal kinase (JNK) pathway and the Drosophila protein Draper (Drpr), characteristic of other neural injury paradigms. We show that macrophages are recruited to the injury site and are required for functional recovery. Further, we show that the proteins Grindelwald and Drpr in Drosophila glial cells mediate activation of JNK, and that expression of drpr is dependent on JNK activation. Finally, we link neuron-glial communication and the requirement of neuronal vesicular transport to regulation of the JNK pathway and functional recovery. This article has an associated First Person interview with the first author of the paper.


Assuntos
Envelhecimento/fisiologia , Sistema Nervoso Central/lesões , Sistema Nervoso Central/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Animais , Sistema Nervoso Central/fisiopatologia , Proteínas de Drosophila/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Compressão Nervosa , Neuroglia/metabolismo , Neurônios/metabolismo , Fagócitos/metabolismo , Recuperação de Função Fisiológica , Vesículas Transportadoras/metabolismo
3.
Int J Mol Sci ; 22(8)2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918666

RESUMO

The c-Jun N-terminal kinase (JNK) signalling pathway is a conserved response to a wide range of internal and external cellular stress signals. Beside the stress response, the JNK pathway is involved in a series of vital regulatory mechanisms during development and adulthood that are critical to maintain tissue homeostasis. These mechanisms include the regulation of apoptosis, growth, proliferation, differentiation, migration and invasion. The JNK pathway has a diverse functionality and cell-tissue specificity, and has emerged as a key player in regeneration, tumorigenesis and other pathologies. The JNK pathway is highly active in the central nervous system (CNS), and plays a central role when cells need to cope with pathophysiological insults during development and adulthood. Here, we review the implications of the JNK pathway in pathologies of the CNS. More specifically, we discuss some newly identified examples and mechanisms of JNK-driven tumor progression in glioblastoma, regeneration/repair after an injury, neurodegeneration and neuronal cell death. All these new discoveries support the central role of JNK in CNS pathologies and reinforce the idea of JNK as potential target to reduce their detrimental effects.


Assuntos
Doenças do Sistema Nervoso Central/etiologia , Doenças do Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/metabolismo , Suscetibilidade a Doenças , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Biomarcadores , Doenças do Sistema Nervoso Central/diagnóstico , Humanos
4.
Elife ; 102021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33527895

RESUMO

Understanding how injury to the central nervous system induces de novo neurogenesis in animals would help promote regeneration in humans. Regenerative neurogenesis could originate from glia and glial neuron-glia antigen-2 (NG2) may sense injury-induced neuronal signals, but these are unknown. Here, we used Drosophila to search for genes functionally related to the NG2 homologue kon-tiki (kon), and identified Islet Antigen-2 (Ia-2), required in neurons for insulin secretion. Both loss and over-expression of ia-2 induced neural stem cell gene expression, injury increased ia-2 expression and induced ectopic neural stem cells. Using genetic analysis and lineage tracing, we demonstrate that Ia-2 and Kon regulate Drosophila insulin-like peptide 6 (Dilp-6) to induce glial proliferation and neural stem cells from glia. Ectopic neural stem cells can divide, and limited de novo neurogenesis could be traced back to glial cells. Altogether, Ia-2 and Dilp-6 drive a neuron-glia relay that restores glia and reprogrammes glia into neural stem cells for regeneration.


Assuntos
Sistema Nervoso Central/lesões , Drosophila melanogaster/crescimento & desenvolvimento , Neurogênese , Regeneração , Animais , Autoanticorpos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Larva/genética , Larva/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Somatomedinas/metabolismo
5.
Int J Dev Biol ; 65(7-8-9): 457-464, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33501996

RESUMO

The Spanish Society for Developmental Biology (SEBD) organized its 17th meeting in November 2020 (herein referred to as SEBD2020). This meeting, originally programmed to take place in the city of Bilbao, was forced onto an online format due to the SARS-CoV2, COVID-19 pandemic. Although, we missed the live personal interactions and missed out on the Bilbao social scene, we were able to meet online to present our work and discuss our latest results. An overview of the activities that took place around the meeting, the different scientific sessions and the speakers involved are presented here. The pros and cons of virtual meetings are discussed.


Assuntos
Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências , Animais , Biologia Celular/tendências , Biologia do Desenvolvimento/educação , Humanos , Internet , Modelos Animais , Sistema Nervoso , Revisão por Pares , Publicações , Editoração , Regeneração , Instituições Acadêmicas , Sociedades Médicas , Espanha
6.
PLoS Biol ; 17(12): e3000545, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31846454

RESUMO

Glioblastoma (GB) is the most lethal brain tumor, and Wingless (Wg)-related integration site (WNT) pathway activation in these tumors is associated with a poor prognosis. Clinically, the disease is characterized by progressive neurological deficits. However, whether these symptoms result from direct or indirect damage to neurons is still unresolved. Using Drosophila and primary xenografts as models of human GB, we describe, here, a mechanism that leads to activation of WNT signaling (Wg in Drosophila) in tumor cells. GB cells display a network of tumor microtubes (TMs) that enwrap neurons, accumulate Wg receptor Frizzled1 (Fz1), and, thereby, deplete Wg from neurons, causing neurodegeneration. We have defined this process as "vampirization." Furthermore, GB cells establish a positive feedback loop to promote their expansion, in which the Wg pathway activates cJun N-terminal kinase (JNK) in GB cells, and, in turn, JNK signaling leads to the post-transcriptional up-regulation and accumulation of matrix metalloproteinases (MMPs), which facilitate TMs' infiltration throughout the brain, TMs' network expansion, and further Wg depletion from neurons. Consequently, GB cells proliferate because of the activation of the Wg signaling target, ß-catenin, and neurons degenerate because of Wg signaling extinction. Our findings reveal a molecular mechanism for TM production, infiltration, and maintenance that can explain both neuron-dependent tumor progression and also the neural decay associated with GB.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Metaloproteinases da Matriz/metabolismo , Neurônios/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Animais Geneticamente Modificados , Neoplasias Encefálicas/patologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Receptores Frizzled/metabolismo , Glioblastoma/patologia , Xenoenxertos , Humanos , Masculino , Microtúbulos/metabolismo , Neurônios/patologia , Proteína Wnt1/metabolismo
7.
Evol Psychol ; 17(2): 1474704919852918, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31167552

RESUMO

Bodily attractiveness is an important component of mate value. Musculature-a crucial component of men's bodily attractiveness-provides women with probabilistic information regarding a potential mate's quality. Overall musculature is comprised of several muscle groups, each of which varies in information value; different muscles should be weighted differently by attractiveness-assessment adaptations as a result. In the current study, women and men ( N = 1,742) reported size preferences for 14 major muscle groups. Women's reported preferences provided only partial support for our hypotheses that women will prefer muscles that most reliably differentiate between potential mates to be larger; men tended to prefer larger upper-body muscles. We discuss possible interpretations of these mixed findings. Ultimately, our findings suggest that attractiveness-assessment adaptations are sensitive to the information contained within specific muscle groups and they highlight the potential for additional research on the nuances of bodily attractiveness assessment.


Assuntos
Beleza , Composição Corporal/fisiologia , Homens , Músculo Esquelético , Aptidão Física/psicologia , Parceiros Sexuais , Adolescente , Adulto , Comportamento de Escolha/fisiologia , Feminino , Humanos , Masculino , Adulto Jovem
8.
J Neurogenet ; 32(2): 78-91, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29718753

RESUMO

In the last years, glial cells have emerged as central players in the development and function of complex nervous systems. Therefore, the concept of glial cells has evolved from simple supporting cells to essential actors. The molecular mechanisms that govern glial functions are evolutionarily conserved from Drosophila to mammals, highlighting genetic similarities between these groups, as well as the great potential of Drosophila research for the understanding of human CNS. These similarities would imply a common phylogenetic origin of glia, even though there is a controversy at this point. This review addresses the existing literature on the evolutionary origin of glia and discusses whether or not insect and mammalian glia are homologous or analogous. Besides, this manuscript summarizes the main glial functions in the CNS and underscores the evolutionarily conserved molecular mechanisms between Drosophila and mammals. Finally, I also consider the current nomenclature and classification of glial cells to highlight the need for a consensus agreement and I propose an alternative nomenclature based on function that unifies Drosophila and mammalian glial types.


Assuntos
Evolução Biológica , Sistema Nervoso Central/citologia , Sistema Nervoso Central/fisiologia , Neuroglia/classificação , Neuroglia/citologia , Neuroglia/fisiologia , Animais , Drosophila , Humanos , Mamíferos , Filogenia
9.
PLoS One ; 13(4): e0194281, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29634720

RESUMO

In this study, we identify the means by which segmentally homologous neurons acquire different neuropeptide fates in Drosophila. Ventral abdominal (Va)-neurons in the A1 segment of the ventral nerve cord express DH31 and AstA neuropeptides (neuropeptidergic fate I) by virtue of Ubx activity, whereas the A2-A4 Va-neurons express the Capa neuropeptide (neuropeptidergic fate II) under the influence of abdA. These different fates are attained through segment-specific programs of neural subtype specification undergone by segmentally homologous neurons. This is an attractive alternative by which Hox genes can shape Drosophila segmental neural architecture (more sophisticated than the previously identified binary "to live" or "not to live" mechanism). These data refine our knowledge of the mechanisms involved in diversifying neuronal identity within the central nervous system.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Hormônios de Inseto/metabolismo , Sistema Nervoso/embriologia , Neuropeptídeos/metabolismo , Oligopeptídeos/metabolismo , Animais , Padronização Corporal/genética , Linhagem da Célula , Sistema Nervoso Central/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Masculino , Sistema Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Temperatura , Fatores de Transcrição/metabolismo
10.
Dev Dyn ; 247(1): 85-93, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28791751

RESUMO

Although the central nervous system does not regenerate, injury induces repair and regenerative responses in glial cells. In mammals, activated microglia clear up apoptotic cells and debris resulting from the injury, astrocytes form a scar that contains the lesion, and NG2-glia elicit a prominent regenerative response. NG2-glia regenerate themselves and differentiate into oligodendrocytes, which remyelinate axons leading to some recovery of locomotion. The regenerative response of glial cells is evolutionarily conserved across the animals and Drosophila genetics revealed an underlying gene network. This involves the genes Notch, kon-tiki, eiger, dorsal, and prospero, homologues of mammalian Notch1, ng2, TNF, NFκB, and prox1, respectively. Feedback loops between these genes enable a surge in proliferation in response to injury and ensuing differentiation. Negative feedback sets a timer for proliferation, and prevents uncontrolled growth that could lead to glioma. Remarkable parallels are found in these genetic relationships between fruit flies and mammals. Drosophila findings provide insights into gene functions that could be manipulated in stem cells and progenitors for therapeutic repair. Developmental Dynamics 247:85-93, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Lesões Encefálicas/fisiopatologia , Redes Reguladoras de Genes , Neuroglia/fisiologia , Regeneração/genética , Traumatismos da Medula Espinal/fisiopatologia , Animais , Lesões Encefálicas/genética , Traumatismos da Medula Espinal/genética
11.
J Cell Biol ; 216(5): 1421-1438, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28373203

RESUMO

Cell number plasticity is coupled to circuitry in the nervous system, adjusting cell mass to functional requirements. In mammals, this is achieved by neurotrophin (NT) ligands, which promote cell survival via their Trk and p75NTR receptors and cell death via p75NTR and Sortilin. Drosophila NTs (DNTs) bind Toll receptors instead to promote neuronal survival, but whether they can also regulate cell death is unknown. In this study, we show that DNTs and Tolls can switch from promoting cell survival to death in the central nervous system (CNS) via a three-tier mechanism. First, DNT cleavage patterns result in alternative signaling outcomes. Second, different Tolls can preferentially promote cell survival or death. Third, distinct adaptors downstream of Tolls can drive either apoptosis or cell survival. Toll-6 promotes cell survival via MyD88-NF-κB and cell death via Wek-Sarm-JNK. The distribution of adaptors changes in space and time and may segregate to distinct neural circuits. This novel mechanism for CNS cell plasticity may operate in wider contexts.


Assuntos
Fatores de Crescimento Neural/metabolismo , Receptores Toll-Like/metabolismo , Animais , Contagem de Células , Morte Celular , Sobrevivência Celular , Drosophila , Plasticidade Neuronal , Transdução de Sinais
12.
Neural Regen Res ; 12(1): 31-34, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28250735

RESUMO

The glial regenerative response to central nervous system (CNS) injury, although limited, can be harnessed to promote regeneration and repair. Injury provokes the proliferation of ensheathing glial cells, which can differentiate to remyelinate axons, and partially restore function. This response is evolutionarily conserved, strongly implying an underlying genetic mechanism. In mammals, it is elicited by NG2 glia, but most often newly generated cells fail to differentiate. Thus an important goal had been to find out how to promote glial differentiation following the proliferative response. A gene network involving Notch and prospero (pros) controls the balance between glial proliferation and differentiation in flies and mice, and promotes CNS repair at least in fruit-flies. A key missing link had been how to relate the function of NG2 to this gene network. Recent findings by Losada-Perez et al., published in JCB, demonstrated that the Drosophila NG2 homologue kon-tiki (kon) is functionally linked to Notch and pros in glia. By engaging in two feedback loops with Notch and Pros, in response to injury, Kon can regulate both glial cell number and glial shape homeostasis, essential for repair. Drosophila offers powerful genetics to unravel the control of stem and progenitor cells for regeneration and repair.

13.
J Cell Biol ; 214(5): 587-601, 2016 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-27551055

RESUMO

Neuron glia antigen 2 (NG2)-positive glia are repair cells that proliferate upon central nervous system (CNS) damage, promoting functional recovery. However, repair is limited because of the failure of the newly produced glial cells to differentiate. It is a key goal to discover how to regulate NG2 to enable glial proliferation and differentiation conducive to repair. Drosophila has an NG2 homologue called kon-tiki (kon), of unknown CNS function. We show that kon promotes repair and identify the underlying mechanism. Crush injury up-regulates kon expression downstream of Notch. Kon in turn induces glial proliferation and initiates glial differentiation by activating glial genes and prospero (pros). Two negative feedback loops with Notch and Pros allow Kon to drive the homeostatic regulation required for repair. By modulating Kon levels in glia, we could prevent or promote CNS repair. Thus, the functional links between Kon, Notch, and Pros are essential for, and can drive, repair. Analogous mechanisms could promote CNS repair in mammals.


Assuntos
Antígenos/química , Sistema Nervoso Central/patologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteoglicanas/química , Homologia de Sequência de Aminoácidos , Cicatrização , Animais , Antígenos/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Forma Celular , Sistema Nervoso Central/metabolismo , Drosophila melanogaster/citologia , Larva/citologia , Larva/metabolismo , Modelos Biológicos , Compressão Nervosa , Neuroglia/citologia , Neuroglia/metabolismo , Proteoglicanas/metabolismo , Regeneração
14.
Cell Tissue Res ; 358(2): 621-6, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25129108

RESUMO

Studies in the Drosophila embryonic NB4-2 lineage have suggested that the transcription factor Klumpfuss (Klu) functions within embryonic neuroblast lineages to differentiate between the identities of two adjacent ganglion mother cells (GMCs). However, because of the limited lineage markers available, these observations have been made only for the NB4-2 lineage. Recent findings have placed this transcription factor in the vanguard of Drosophila neural stem cell biology by demonstrating that Klu is necessary for larval neuroblast growth and self-renewal. Here, we have studied the role of klu in an incipient model in order to address basic mechanisms of neural specification: the Va system. None of the previously reported roles of Klu satisfactorily explain our observations. Unexpectedly, in this lineage, klu is necessary for differentiating between the fates of the two neurons born from a unique GMC; klu mutants produce two B-type cells, rather than one B-type (Notch-OFF) and one A-type (Notch-ON) cell. Additionally, our results demonstrate that Klu operates in the GMC and/or in the newly born neuron, but not in the neuroblast. Unlike in larval neuroblasts in which Klu is an executor of Notch signaling, we have found that Klu does not lie downstream of the Notch pathway in this cell division context.


Assuntos
Divisão Celular Assimétrica , Linhagem da Célula , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Gânglios dos Invertebrados/citologia , Gânglios dos Invertebrados/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
15.
An. psicol ; 30(2): 667-675, mayo 2014. ilus, tab
Artigo em Inglês | IBECS | ID: ibc-121805

RESUMO

A recent and controversial hypothesis suggests the presence of an oestrus phase in women as in other mammals. This implies that women at their optimal fertility point of the menstrual cycle exhibit behaviors focused to maximize the genetic quality of their offspring. Several studies support this hypothesis, finding that women in the fertile phase tend to prefer men with traits associated to phenotypic quality, such as greater facial masculinization and symmetry. We experimentally tested some of the observations supporting this hypothesis in a population of 810 young Spanish women. We analyzed whether the preference for masculinized male faces is affected by I) the phase of the menstrual cycle, II) having a stable partner and III) the use of birth control pills. We could not repro-duce the effect of the first two factors, but we found that women using hormonal contraceptives tend to prefer men with less masculine faces. These results indicate that some of the evidences supporting the oestrus hypothesis in humans must be reviewed, incorporating data from different socio-cultural and ethnic populations


Recientemente se ha postulado una controvertida hipótesis que propone la presencia de un periodo de estro en las mujeres, como ocurre en otros mamíferos. Ello implica que las mujeres en el óptimo de fertilidad del ciclo menstrual presenten comportamientos encaminados a maximizar la calidad genética de su descendencia. Diversas investigaciones sostienen esta hipótesis, al encontrar que las mujeres en la fase fértil prefieren hombres con rasgos que denotan mayor calidad fenotípica, como un mayor grado de masculinización o una mayor simetría. Nuestro objetivo ha sido testar experimentalmente en una población de 810 jóvenes españolas alguna de estas observaciones. Analizamos si, tal como se recoge en la bibliografía, la preferencia por rostros de hombres masculinizados se ve afectada por I) la etapa del ciclo menstrual, II) el tener pareja estable y III) el empleo anticonceptivos hormonales. No hemos podido reproducir el efecto de los dos primeros factores, pero sí encontramos que las mujeres que emplean anticonceptivos hormonales prefirieron rostros de hombre menos masculinos. Estos resultados no refutan la hipótesis del estro en humanos, pero nos indican que algunas de las pruebas que la sustentan han de ser reconsideradas, incorporando datos de poblaciones étnica y socioculturalmente diferentes


Assuntos
Humanos , Feminino , Estro , Comportamento Reprodutivo/psicologia , Testes Psicológicos/estatística & dados numéricos , Caracteres Sexuais , Comportamento Sexual/psicologia , Ciclo Menstrual/psicologia
16.
Development ; 140(10): 2181-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23633512

RESUMO

A number of transcription factors that are expressed within most, if not all, embryonic neuroblast (NB) lineages participate in neural subtype specification. Some have been extensively studied in several NB lineages (e.g. components of the temporal gene cascade) whereas others only within specific NB lineages. To what extent they function in other lineages remains unknown. Klumpfuss (Klu), the Drosophila ortholog of the mammalian Wilms tumor 1 (WT1) protein, is one such transcription factor. Studies in the NB4-2 lineage have suggested that Klu functions to ensure that the two ganglion mother cells (GMCs) in this embryonic NB lineage acquire different fates. Owing to limited lineage marker availability, these observations were made only for the NB4-2 lineage. Recent findings reveal that Klu is necessary for larval neuroblast growth and self-renewal. We have extended the study of Klu to the well-known embryonic NB5-6T lineage and describe a novel role for Klu in the Drosophila embryonic CNS. Our results demonstrate that Klu is expressed specifically in the postmitotic Ap4/FMRFa neuron, promoting its differentiation through the initiation of BMP signaling. Our findings indicate a pleiotropic function of Klu in Ap cluster specification in general and particularly in Ap4 neuron differentiation, indicating that Klu is a multitasking transcription factor. Finally, our studies indicate that a transitory downregulation of klu is crucial for the specification of the Ap4/FMRFa neuron. Similar to WT1, klu seems to have either self-renewal or differentiation-promoting functions, depending on the developmental context.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , FMRFamida/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Análise por Conglomerados , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Microscopia Confocal , Neurônios/metabolismo , Transdução de Sinais
17.
Mech Dev ; 128(3-4): 208-21, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21236339

RESUMO

The central nervous system contains a wide variety of neuronal subclasses generated by neural progenitors. The achievement of a unique neural fate is the consequence of a sequence of early and increasingly restricted regulatory events, which culminates in the expression of a specific genetic combinatorial code that confers individual characteristics to the differentiated cell. How the earlier regulatory events influence post-mitotic cell fate decisions is beginning to be understood in the Drosophila NB 5-6 lineage. However, it remains unknown to what extent these events operate in other lineages. To better understand this issue, we have used a very highly specific marker that identifies a small subset of abdominal cells expressing the Drosophila neuropeptide Capa: the ABCA neurons. Our data support the birth of the ABCA neurons from NB 5-3 in a cas temporal window in the abdominal segments A2-A4. Moreover, we show that the ABCA neuron has an ABCA-sibling cell which dies by apoptosis. Surprisingly, both cells are also generated in the abdominal segments A5-A7, although they undergo apoptosis before expressing Capa. In addition, we have performed a targeted genetic screen to identify players involved in ABCA specification. We have found that the ABCA fate requires zfh2, grain, Grunge and hedgehog genes. Finally, we show that the NB 5-3 generates other subtype of Capa-expressing cells (SECAs) in the third suboesophageal segment, which are born during a pdm/cas temporal window, and have different genetic requirements for their specification.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Abdome/inervação , Animais , Antígenos de Diferenciação/metabolismo , Padronização Corporal/genética , Morte Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Proteínas Hedgehog/metabolismo , Tecido Nervoso/citologia , Tecido Nervoso/embriologia , Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neuropeptídeos/genética , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
18.
Mech Dev ; 127(9-12): 458-71, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20732418

RESUMO

It is becoming increasingly clear that the activation of specific terminal differentiation genes during neural development is critically dependent upon the establishment of unique combinatorial transcription factor codes within distinct neural cell subtypes. However, it is still unclear to which extent these codes are shared by lineage-unrelated neurons expressing the same terminal differentiation genes. Additionally, it is not known if the activation of a specific terminal differentiation gene is restricted to cells born at a particular developmental time point. Here, we utilize the terminal differentiation gene FMRFa which is expressed by the Ap4 and SE2 neurons in the Drosophila ventral nerve cord, to explore these issues in depth. We find that the Ap4 and SE2 neurons are generated by different neural progenitors and use different combinatorial codes to activate FMRFa expression. Additionally, we find that the Ap4 and SE2 neurons are generated in different temporal gene expression windows. Extending the investigation to include a second Drosophila terminal differentiation gene, Leucokinin, we find similar results, suggesting that neurons generated by different progenitors might commonly use different transcription factor codes to activate the same terminal differentiation gene. Furthermore, these results imply that the activation of a particular terminal differentiation gene in temporally unrestricted.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/citologia , Neurônios/metabolismo , Animais , Biomarcadores/metabolismo , Padronização Corporal/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , FMRFamida/genética , FMRFamida/metabolismo , Genes de Insetos/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fenótipo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...