Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
BMC Genomics ; 25(1): 570, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844864

RESUMEN

Compound eyes formation in decapod crustaceans occurs after the nauplius stage. However, the key genes and regulatory mechanisms of compound eye development during crustacean embryonic development have not yet been clarified. In this study, RNA-seq was used to investigate the gene expression profiles of Neocaridina denticulata sinensis from nauplius to zoea stage. Based on RNA-seq data analysis, the phototransduction and insect hormone biosynthesis pathways were enriched, and molting-related neuropeptides were highly expressed. There was strong cell proliferation in the embryo prior to compound eye development. The formation of the visual system and the hormonal regulation of hatching were the dominant biological events during compound eye development. The functional analysis of DEGs across all four developmental stages showed that cuticle formation, muscle growth and the establishment of immune system occurred from nauplius to zoea stage. Key genes related to eye development were discovered, including those involved in the determination and differentiation of the eye field, eye-color formation, and visual signal transduction. In conclusion, the results increase the understanding of the molecular mechanism of eye formation in crustacean embryonic stage.


Asunto(s)
Ojo Compuesto de los Artrópodos , Perfilación de la Expresión Génica , Animales , Ojo Compuesto de los Artrópodos/metabolismo , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Transcriptoma , Regulación del Desarrollo de la Expresión Génica , Decápodos/genética , Decápodos/crecimiento & desarrollo , Ojo/metabolismo , Ojo/embriología , Ojo/crecimiento & desarrollo
2.
Cell Death Dis ; 13(2): 101, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110540

RESUMEN

Hippo signaling is a conserved mechanism for controlling organ growth. Increasing evidence suggests that Hippo signaling is modulated by various cellular factors for normal development and tumorigenesis. Hence, identification of these factors is pivotal for understanding the mechanism for the regulation of Hippo signaling. Drosophila Mnat9 is a putative N-acetyltransferase that is required for cell survival by affecting JNK signaling. Here we show that Mnat9 is involved in the negative regulation of Hippo signaling. RNAi knockdown of Mnat9 in the eye disc suppresses the rough eye phenotype of overexpressing Crumbs (Crb), an upstream factor of the Hippo pathway. Conversely, Mnat9 RNAi enhances the eye phenotype caused by overexpressing Expanded (Ex) or Warts (Wts) that acts downstream to Crb. Similar genetic interactions between Mnat9 and Hippo pathway genes are found in the wing. The reduced wing phenotype of Mnat9 RNAi is suppressed by overexpression of Yorkie (Yki), while it is suppressed by knockdown of Hippo upstream factors like Ex, Merlin, or Kibra. Mnat9 co-immunoprecipitates with Mer, implying their function in a protein complex. Furthermore, Mnat9 overexpression together with Hpo knockdown causes tumorous overgrowth in the abdomen. Our data suggest that Mnat9 is required for organ growth and can induce tumorous growth by negatively regulating the Hippo signaling pathway.


Asunto(s)
Carcinogénesis/metabolismo , Drosophila melanogaster/metabolismo , Vía de Señalización Hippo , Acetiltransferasas N-Terminal/metabolismo , Animales , Carcinogénesis/patología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Acetiltransferasas N-Terminal/genética , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño , Alas de Animales/metabolismo
3.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35072208

RESUMEN

The eye-antennal disc of Drosophila is composed of three cell layers: a columnar epithelium called the disc proper (DP); an overlying sheet of squamous cells called the peripodial epithelium (PE); and a strip of cuboidal cells that joins the other two cellular sheets to each other and comprises the outer margin (M) of the disc. The M cells play an important role in patterning the eye because it is here that the Hedgehog (Hh), Decapentaplegic (Dpp) and JAK/STAT pathways function to initiate pattern formation. Dpp signaling is lost from the margin of eyes absent (eya) mutant discs and, as a result, the initiation of retinal patterning is blocked. Based on these observations, Eya has been proposed to control the initiation of the morphogenetic furrow via regulation of Dpp signaling within the M. We show that the failure in pattern formation surprisingly results from M cells prematurely adopting a head epidermis fate. This switch in fate normally takes place during pupal development after the eye has been patterned. Our results suggest that the timing of cell fate decisions is essential for correct eye development.


Asunto(s)
Ojo Compuesto de los Artrópodos/citología , Proteínas de Drosophila/metabolismo , Proteínas del Ojo/metabolismo , Animales , Diferenciación Celular , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas del Ojo/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Quinasas Janus/metabolismo , Morfogénesis , Mutación , Factores de Transcripción STAT/metabolismo
4.
PLoS Biol ; 19(8): e3001367, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34379617

RESUMEN

Damage in the nervous system induces a stereotypical response that is mediated by glial cells. Here, we use the eye disc of Drosophila melanogaster as a model to explore the mechanisms involved in promoting glial cell response after neuronal cell death induction. We demonstrate that these cells rapidly respond to neuronal apoptosis by increasing in number and undergoing morphological changes, which will ultimately grant them phagocytic abilities. We found that this glial response is controlled by the activity of Decapentaplegic (Dpp) and Hedgehog (Hh) signalling pathways. These pathways are activated after cell death induction, and their functions are necessary to induce glial cell proliferation and migration to the eye discs. The latter of these 2 processes depend on the function of the c-Jun N-terminal kinase (JNK) pathway, which is activated by Dpp signalling. We also present evidence that a similar mechanism controls glial response upon apoptosis induction in the leg discs, suggesting that our results uncover a mechanism that might be involved in controlling glial cells response to neuronal cell death in different regions of the peripheral nervous system (PNS).


Asunto(s)
Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Proteínas Hedgehog/fisiología , Neuroglía/fisiología , Animales , Apoptosis , Movimiento Celular , Ojo Compuesto de los Artrópodos/citología , Drosophila melanogaster/citología , Sistema de Señalización de MAP Quinasas
5.
Dev Biol ; 478: 173-182, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34245727

RESUMEN

A complex network of transcription factor interactions propagates across the larval eye disc to establish columns of evenly-spaced R8 precursor cells, the founding cells of Drosophila ommatidia. After the recruitment of additional photoreceptors to each ommatidium, the surrounding cells are organized into their stereotypical pattern during pupal development. These support cells - comprised of pigment and cone cells - are patterned to encapsulate the photoreceptors and separate ommatidia with an hexagonal honeycomb lattice. Since the proteins and processes essential for correct eye patterning are conserved, elucidating how these function and change during Drosophila eye patterning can substantially advance our understanding of transcription factor and signaling networks, cytoskeletal structures, adhesion complexes, and the biophysical properties of complex tissues during their morphogenesis. Our understanding of many of these aspects of Drosophila eye patterning is largely descriptive. Many important questions, especially relating to the regulation and integration of cellular events, remain.


Asunto(s)
Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Drosophila/crecimiento & desarrollo , Células Fotorreceptoras de Invertebrados/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo , Ojo Compuesto de los Artrópodos/citología , Simulación por Computador , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores ErbB/metabolismo , Larva/crecimiento & desarrollo , Morfogénesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras de Invertebrados/citología , Pupa/crecimiento & desarrollo , Receptores de Péptidos de Invertebrados/metabolismo , Transducción de Señal
6.
Genetics ; 217(4)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33681970

RESUMEN

In the last larval instar, uncommitted progenitor cells in the Drosophila eye primordium start to adopt individual retinal cell fates, arrest their growth and proliferation, and initiate terminal differentiation into photoreceptor neurons and other retinal cell types. To explore the regulation of these processes, we have performed mRNA-Seq studies of the larval eye and antennal primordial at multiple developmental stages. A total of 10,893 fly genes were expressed during these stages and could be adaptively clustered into gene groups, some of whose expression increases or decreases in parallel with the cessation of proliferation and onset of differentiation. Using in situ hybridization of a sample of 98 genes to verify spatial and temporal expression patterns, we estimate that 534 genes or more are transcriptionally upregulated during retinal differentiation, and 1367 or more downregulated as progenitor cells differentiate. Each group of co-expressed genes is enriched for regulatory motifs recognized by co-expressed transcription factors, suggesting that they represent coherent transcriptional regulatory programs. Using available mutant strains, we describe novel roles for the transcription factors SoxNeuro (SoxN), H6-like homeobox (Hmx), CG10253, without children (woc), Structure specific recognition protein (Ssrp), and multisex combs (mxc).


Asunto(s)
Ojo Compuesto de los Artrópodos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transcriptoma , Animales , Diferenciación Celular , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Sci Rep ; 11(1): 1111, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441820

RESUMEN

Human papillomavirus (HPV) is the leading cause of cervical cancer and has been implicated in several other cancer types including vaginal, vulvar, penile, and oropharyngeal cancers. Despite the recent availability of a vaccine, there are still over 310,000 deaths each year worldwide. Current treatments for HPV-mediated cancers show limited efficacy, and would benefit from improved understanding of disease mechanisms. Recently, we developed a Drosophila 'HPV 18 E6' model that displayed loss of cellular morphology and polarity, junctional disorganization, and degradation of the major E6 target Magi; we further provided evidence that mechanisms underlying HPV E6-induced cellular abnormalities are conserved between humans and flies. Here, we report a functional genetic screen of the Drosophila kinome that identified IKK[Formula: see text]-a regulator of NF-κB-as an enhancer of E6-induced cellular defects. We demonstrate that inhibition of IKK[Formula: see text] reduces Magi degradation and that this effect correlates with hyperphosphorylation of E6. Further, the reduction in IKK[Formula: see text] suppressed the cellular transformation caused by the cooperative action of HPVE6 and the oncogenic Ras. Finally, we demonstrate that the interaction between IKK[Formula: see text] and E6 is conserved in human cells: inhibition of IKK[Formula: see text] blocked the growth of cervical cancer cells, suggesting that IKK[Formula: see text] may serve as a novel therapeutic target for HPV-mediated cancers.


Asunto(s)
Ojo Compuesto de los Artrópodos/anomalías , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Viral , Ojo Compuesto de los Artrópodos/citología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Drosophila , Femenino , Humanos , Nucleósido-Fosfato Quinasa/metabolismo , Dominios PDZ , Fosforilación , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo
8.
Mol Biol Evol ; 38(5): 1924-1942, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33386848

RESUMEN

Revealing the mechanisms underlying the breathtaking morphological diversity observed in nature is a major challenge in Biology. It has been established that recurrent mutations in hotspot genes cause the repeated evolution of morphological traits, such as body pigmentation or the gain and loss of structures. To date, however, it remains elusive whether hotspot genes contribute to natural variation in the size and shape of organs. As natural variation in head morphology is pervasive in Drosophila, we studied the molecular and developmental basis of differences in compound eye size and head shape in two closely related Drosophila species. We show differences in the progression of retinal differentiation between species and we applied comparative transcriptomics and chromatin accessibility data to identify the GATA transcription factor Pannier (Pnr) as central factor associated with these differences. Although the genetic manipulation of Pnr affected multiple aspects of dorsal head development, the effect of natural variation is restricted to a subset of the phenotypic space. We present data suggesting that this developmental constraint is caused by the coevolution of expression of pnr and its cofactor u-shaped (ush). We propose that natural variation in expression or function of highly connected developmental regulators with pleiotropic functions is a major driver for morphological evolution and we discuss implications on gene regulatory network evolution. In comparison to previous findings, our data strongly suggest that evolutionary hotspots are not the only contributors to the repeated evolution of eye size and head shape in Drosophila.


Asunto(s)
Evolución Biológica , Ojo Compuesto de los Artrópodos/anatomía & histología , Drosophila/anatomía & histología , Pleiotropía Genética , Animales , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Drosophila/fisiología , Proteínas de Drosophila/metabolismo , Femenino , Redes Reguladoras de Genes , Cabeza/anatomía & histología , Larva/crecimiento & desarrollo , Masculino , Especificidad de la Especie , Factores de Transcripción/metabolismo , Transcriptoma
9.
Genesis ; 58(10-11): e23395, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32990387

RESUMEN

During organogenesis, cell proliferation is followed by the differentiation of specific cell types to form an organ. Any aberration in differentiation can result in developmental defects, which can result in a partial to a near-complete loss of an organ. We employ the Drosophila eye model to understand the genetic and molecular mechanisms involved in the process of differentiation. In a forward genetic screen, we identified, cullin-4 (cul-4), which encodes an E3 ubiquitin ligase, to play an important role in retinal differentiation. During development, cul-4 is known to be involved in protein degradation, regulation of genomic stability, and regulation of cell cycle. Previously, we have reported that cul-4 regulates cell death during eye development by downregulating Wingless (Wg)/Wnt signaling pathway. We found that loss-of-function of cul-4 results in a reduced eye phenotype, which can be due to onset of cell death. However, we found that loss-of-function of cul-4 also affects retinal development by downregulating retinal determination (RD) gene expression. Early markers of retinal differentiation are dysregulated in cul-4 loss of function conditions, indicating that cul-4 is necessary for differentiation. Furthermore, loss-of-function of cul-4 ectopically induces expression of negative regulators of eye development like Wg and Homothorax (Hth). During eye development, Wg is known to block the progression of a synchronous wave of differentiation referred to as Morphogenetic furrow (MF). In cul-4 loss-of-function background, expression of dpp-lacZ, a MF marker, is significantly downregulated. Our data suggest a new role of cul-4 in retinal differentiation. These studies may have significant bearings on our understanding of early eye development.


Asunto(s)
Ojo Compuesto de los Artrópodos/metabolismo , Proteínas Cullin/metabolismo , Proteínas de Drosophila/metabolismo , Neurogénesis , Animales , Ojo Compuesto de los Artrópodos/citología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas Cullin/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulación del Desarrollo de la Expresión Génica , Mutación con Pérdida de Función , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Vía de Señalización Wnt
10.
Genetics ; 215(1): 117-128, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32122936

RESUMEN

The Hippo pathway is an evolutionarily conserved signaling network that regulates organ size, cell fate, and tumorigenesis. In the context of organ size control, the pathway incorporates a large variety of cellular cues, such as cell polarity and adhesion, into an integrated transcriptional response. The central Hippo signaling effector is the transcriptional coactivator Yorkie, which controls gene expression in partnership with different transcription factors, most notably Scalloped. When it is not activated by Yorkie, Scalloped can act as a repressor of transcription, at least in part due to its interaction with the corepressor protein Tgi. The mechanism by which Tgi represses transcription is incompletely understood, and therefore we sought to identify proteins that potentially operate together with Tgi. Using an affinity purification and mass-spectrometry approach we identified Pits and CtBP as Tgi-interacting proteins, both of which have been linked to transcriptional repression. Both Pits and CtBP were required for Tgi to suppress the growth of the Drosophila melanogaster eye and CtBP loss suppressed the undergrowth of yorkie mutant eye tissue. Furthermore, as reported previously for Tgi, overexpression of Pits repressed transcription of Hippo pathway target genes. These findings suggest that Tgi might operate together with Pits and CtBP to repress transcription of genes that normally promote tissue growth. The human orthologs of Tgi, CtBP, and Pits (VGLL4, CTBP2, and IRF2BP2) have previously been shown to physically and functionally interact to control transcription, implying that the mechanism by which these proteins control transcriptional repression is conserved throughout evolution.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Proteínas Portadoras/metabolismo , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/metabolismo , Oxidorreductasas de Alcohol/genética , Animales , Proteínas Portadoras/genética , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas Nucleares/genética , Unión Proteica , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Señalizadoras YAP
11.
Dev Cell ; 52(5): 605-616.e7, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32032548

RESUMEN

The expression of multiple growth-promoting genes is coordinated by the transcriptional co-activator Yorkie with its major regulatory input provided by the Hippo-Warts kinase cascade. Here, we identify Atg1/ULK1-mediated phosphorylation of Yorkie as an additional inhibitory input independent of the Hippo-Warts pathway. Two serine residues in Yorkie, S74 and S97, are Atg1/ULK1 consensus target sites and are phosphorylated by ULK1 in vitro, thereby preventing its binding to Scalloped. In vivo, gain of function of Atg1, or its activator Acinus, caused elevated Yorkie phosphorylation and inhibited Yorkie's growth-promoting activity. Loss of function of Atg1 or Acinus raised expression of Yorkie target genes and increased tissue size. Unlike Atg1's role in autophagy, Atg1-mediated phosphorylation of Yorkie does not require Atg13. Atg1 is activated by starvation and other cellular stressors and therefore can impose temporary stress-induced constraints on the growth-promoting gene networks under the control of Hippo-Yorkie signaling.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Sitios de Unión , Núcleo Celular/metabolismo , Ojo Compuesto de los Artrópodos/metabolismo , Secuencia de Consenso , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Transactivadores/química , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
12.
Mol Brain ; 13(1): 1, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31900209

RESUMEN

The establishment of the functional nervous system requires coordinated development of neurons and glia in the embryo. Our understanding of underlying molecular and cellular mechanisms, however, remains limited. The developing Drosophila visual system is an excellent model for understanding the developmental control of the nervous system. By performing a systematic transgenic RNAi screen, we investigated the requirements of secreted proteins and cell-surface receptors for the development of photoreceptor neurons (R cells) and wrapping glia (WG) in the Drosophila visual system. From the screen, we identified seven genes whose knockdown disrupted the development of R cells and/or WG, including amalgam (ama), domeless (dome), epidermal growth factor receptor (EGFR), kuzbanian (kuz), N-Cadherin (CadN), neuroglian (nrg), and shotgun (shg). Cell-type-specific analysis revealed that ama is required in the developing eye disc for promoting cell proliferation and differentiation, which is essential for the migration of glia in the optic stalk. Our results also suggest that nrg functions in both eye disc and WG for coordinating R-cell and WG development.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Discos Imaginales/metabolismo , Inmunoglobulinas/fisiología , Neurogénesis/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Interferencia de ARN , Animales , Animales Modificados Genéticamente , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Linaje de la Célula , Movimiento Celular , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Técnicas de Silenciamiento del Gen , Discos Imaginales/citología , Inmunoglobulinas/genética , Larva , Células Fotorreceptoras de Invertebrados/citología , Células Fotorreceptoras de Invertebrados/metabolismo
13.
G3 (Bethesda) ; 10(1): 57-67, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31704710

RESUMEN

Phosphoinositides are lipid signaling molecules that regulate several conserved sub-cellular processes in eukaryotes, including cell growth. Phosphoinositides are generated by the enzymatic activity of highly specific lipid kinases and phosphatases. For example, the lipid PIP3, the Class I PI3 kinase that generates it and the phosphatase PTEN that metabolizes it are all established regulators of growth control in metazoans. To identify additional functions for phosphoinositides in growth control, we performed a genetic screen to identify proteins which when depleted result in altered tissue growth. By using RNA-interference mediated depletion coupled with mosaic analysis in developing eyes, we identified and classified additional candidates in the developing Drosophila melanogaster eye that regulate growth either cell autonomously or via cell-cell interactions. We report three genes: Pi3K68D, Vps34 and fwd that are important for growth regulation and suggest that these are likely to act via cell-cell interactions in the developing eye. Our findings define new avenues for the understanding of growth regulation in metazoan tissue development by phosphoinositide metabolizing proteins.


Asunto(s)
Procesos de Crecimiento Celular/genética , Fosfatidilinositoles/genética , Transducción de Señal , Animales , Fosfatidilinositol 3-Quinasas Clase III/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Ojo Compuesto de los Artrópodos/citología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
14.
Genetics ; 213(3): 877-895, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31558581

RESUMEN

Heterochromatin-mediated repression is essential for controlling the expression of transposons and for coordinated cell type-specific gene regulation. The small ovary (sov) locus was identified in a screen for female-sterile mutations in Drosophila melanogaster, and mutants show dramatic ovarian morphogenesis defects. We show that the null sov phenotype is lethal and map the locus to the uncharacterized gene CG14438, which encodes a nuclear zinc-finger protein that colocalizes with the essential Heterochromatin Protein 1 (HP1a). We demonstrate Sov functions to repress inappropriate gene expression in the ovary, silence transposons, and suppress position-effect variegation in the eye, suggesting a central role in heterochromatin stabilization.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Heterocromatina/metabolismo , Animales , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Elementos Transponibles de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Heterocromatina/genética , Mutación con Pérdida de Función , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Dedos de Zinc
15.
Invert Neurosci ; 19(1): 2, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30603776

RESUMEN

Our study aims to describe (1) external morphology of the compound eye of Antilochus conquebertii, (2) postembryonic changes involving the eye's shape and size and (3) behaviour of the animal with respect to the organization of the compound eye. With each moult of the insect, the structural units of the compound eye increase in size as well as the number, resulting in an overall increase in eye size. The resolution of the adult eye is better than the young one. The adult possesses UV and polarization sensitivity in its eye. Parallel to the changes of the eye the behaviour of the adult animal changes, rendering it increasingly nocturnal and less active in under illuminated conditions. The current study describes the eye and its functional relationship with the behaviour of the animal at the nymphal and adult developmental stage.


Asunto(s)
Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/ultraestructura , Hemípteros/fisiología , Hemípteros/ultraestructura , Visión Ocular/fisiología , Animales , Conducta Alimentaria/fisiología
16.
Dev Genes Evol ; 229(1): 13-24, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30612166

RESUMEN

During development of higher animals, the Notch signalling pathway governs cell type specification by mediating appropriate gene expression responses. In the absence of signalling, Notch target genes are silenced by repressor complexes. In the model organism Drosophila melanogaster, the repressor complex includes the transcription factor Suppressor of Hairless [Su(H)] and Hairless (H) plus general co-repressors. Recent crystal structure analysis of the Drosophila Notch repressor revealed details of the Su(H)-H complex. They were confirmed by mutational analyses of either protein; however, only Su(H) mutants have been further studied in vivo. Here, we analyse three H variants predicted to affect Su(H) binding. To this end, amino acid replacements Phenylalanine 237, Leucines 245 and 247, as well as Tryptophan 258 to Alanine were introduced into the H protein. A cell-based reporter assay indicates substantial loss of Su(H) binding to the respective mutant proteins HFA, HLLAA and HWA. For in vivo analysis, UAS-lines HFA, HLLAA and HWA were generated to allow spatially restricted overexpression. In these assays, all three mutants resembled the HLD control, shown before to lack Su(H) binding, indicating a strong reduction of H activity. For example, the H variants were impaired in wing margin formation, but unexpectedly induced ectopic wing venation. Concurrent overexpression with Su(H), however, suggests that all mutant H protein isoforms are still able to bind Su(H) in vivo. We conclude that a weakening of the cohesion in the H-Su(H) repressor complex is sufficient for disrupting its in vivo functionality.


Asunto(s)
Proteínas de Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Mutación , Receptores Notch/metabolismo , Factores de Transcripción/genética , Animales , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Unión Proteica , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo
17.
Nat Commun ; 9(1): 3626, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30194291

RESUMEN

Haploinsufficiency and aneuploidy are two phenomena, where gene dosage alterations cause severe defects ultimately resulting in developmental failures and disease. One remarkable exception is the X chromosome, where copy number differences between sexes are buffered by dosage compensation systems. In Drosophila, the Male-Specific Lethal complex (MSLc) mediates upregulation of the single male X chromosome. The evolutionary origin and conservation of this process orchestrated by MSL2, the only male-specific protein within the fly MSLc, have remained unclear. Here, we report that MSL2, in addition to regulating the X chromosome, targets autosomal genes involved in patterning and morphogenesis. Precise regulation of these genes by MSL2 is required for proper development. This set of dosage-sensitive genes maintains such regulation during evolution, as MSL2 binds and similarly regulates mouse orthologues via Histone H4 lysine 16 acetylation. We propose that this gene-by-gene dosage compensation mechanism was co-opted during evolution for chromosome-wide regulation of the Drosophila male X.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Compensación de Dosificación (Genética) , Proteínas de Drosophila/fisiología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes del Desarrollo , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Tipificación del Cuerpo , Células Cultivadas , Ensamble y Desensamble de Cromatina , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Drosophila , Femenino , Larva/metabolismo , Masculino , Ratones , Ratones Noqueados , Alas de Animales/crecimiento & desarrollo
18.
Biol Lett ; 14(7)2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30045903

RESUMEN

In insects, the subdivision of the head into a lateral region, harbouring the compound eyes (CEs), and a dorsal (medial) region, where the ocelli localize, is conserved. This organization might have been already present in the insects' euarthropodan ancestors. In Drosophila, the Wnt-1 homologue wingless (wg) plays a major role in the genetic subdivision of the head. To analyse specifically the role of wg signalling in the development of the dorsal head, we attenuated this pathway specifically in this region by genetic means. We find that loss of wg signalling transforms the dorsal/medial head into lateral head structures, including the development of ectopic CEs. Our genetic analysis further suggests that wg signalling organizes the dorsal head medial-lateral axis by controlling, at least in part, the expression domains of the transcription factors Otd and Ey/Pax6.


Asunto(s)
Drosophila/genética , Cabeza/crecimiento & desarrollo , Animales , Tipificación del Cuerpo , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Drosophila/crecimiento & desarrollo , Genes de Insecto/genética , Larva/genética , Larva/crecimiento & desarrollo , Transducción de Señal/genética , Factores de Transcripción/genética
19.
Brain Res ; 1689: 30-44, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29604258

RESUMEN

Mutations in SLC25A46 gene have been identified in mitochondrial diseases that are sometimes classified as Charcot-Marie-Tooth disease type 2, optic atrophy and Leigh syndrome. Human SLC25A46 functions as a transporter across the outer mitochondrial membrane. However, it is still unknown how the neurodegeneration occurring in these diseases relates to the loss of SLC25A46 function. Drosophila has CG5755 (dSLC25A46) as a single human SLC25A46 homolog. Here we established pan-neuron specific dSLC25A46 knockdown flies, and examined their phenotypes. Neuron specific knockdown of dSLC25A46 resulted in an impaired motility in both larvae and adults. Defects at neuromuscular junctions (NMJs), such as reduced synaptic branch length, decreased number and size of bouton, reduced density and size of active zone were also observed with the dSLC25A46 knockdown flies. Mitochondrial hyperfusion in synapse at NMJ, accumulation of reactive oxygen species and reduction of ATP were also observed in the dSLC25A46 knockdown flies. These results indicate that depletion of SLC25A46 induces mitochondrial defects accompanied with aberrant morphology of motoneuron and reduction of active zone that results in defect in locomotive ability. In addition, it is known that SLC25A46 mutations in human cause optic atrophy and knockdown of dSLC25A46 induces aberrant morphology of optic stalk of photoreceptor neurons in third instar larvae. Morphology and development of optic stalk of photoreceptor neurons in Drosophila are precisely regulated via cell proliferation and migration. Immunocytochemical analyses of subcellular localization of dSLC25A46 revealed that dSLC25A46 localizes not only in mitochondria, but also in plasma membrane. These observations suggest that in addition to the role in mitochondrial function, plasma membrane-localized dSLC25A46 plays a role in cell proliferation and/or migration to control optic stalk formation. The dSLC25A46 knockdown fly thus recapitulates most of the phenotypes in mitochondrial disease patients, providing a useful tool to study these diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Enfermedades Mitocondriales , Adenosina Trifosfato/metabolismo , Animales , Animales Modificados Genéticamente , Membrana Celular/metabolismo , Membrana Celular/patología , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Ojo Compuesto de los Artrópodos/patología , Drosophila/genética , Técnicas de Silenciamiento del Gen , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/genética , Actividad Motora/fisiología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Proteínas de Transporte de Fosfato/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Glándulas Salivales/crecimiento & desarrollo , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Homología de Secuencia de Aminoácido
20.
Front Neural Circuits ; 11: 96, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29238294

RESUMEN

Following gene duplication events, the expression patterns of the resulting gene copies can often diverge both spatially and temporally. Here we report on gene duplicates that are expressed in distinct but overlapping patterns, and which exhibit temporally divergent expression. Butterflies have sophisticated color vision and spectrally complex eyes, typically with three types of heterogeneous ommatidia. The eyes of the butterfly Papilio xuthus express two green- and one red-absorbing visual pigment, which came about via gene duplication events, in addition to one ultraviolet (UV)- and one blue-absorbing visual pigment. We localized mRNAs encoding opsins of these visual pigments in developing eye disks throughout the pupal stage. The mRNAs of the UV and blue opsin are expressed early in pupal development (pd), specifying the type of the ommatidium in which they appear. Red sensitive photoreceptors first express a green opsin mRNA, which is replaced later by the red opsin mRNA. Broadband photoreceptors (that coexpress the green and red opsins) first express the green opsin mRNA, later change to red opsin mRNA and finally re-express the green opsin mRNA in addition to the red mRNA. Such a unique temporal and spatial expression pattern of opsin mRNAs may reflect the evolution of visual pigments and provide clues toward understanding how the spectrally complex eyes of butterflies evolved.


Asunto(s)
Mariposas Diurnas/crecimiento & desarrollo , Mariposas Diurnas/metabolismo , Proteínas de Insectos/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Opsinas de Bastones/metabolismo , Animales , Mariposas Diurnas/anatomía & histología , Mariposas Diurnas/ultraestructura , Ojo Compuesto de los Artrópodos/anatomía & histología , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Ojo Compuesto de los Artrópodos/ultraestructura , Femenino , Hibridación in Situ , Microscopía Electrónica , Células Fotorreceptoras de Invertebrados/ultraestructura , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...