Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nucleic Acids Res ; 52(5): 2093-2111, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38303573

RESUMEN

Co-transcriptional processing of nascent pre-mRNAs by the spliceosome is vital to regulating gene expression and maintaining genome integrity. Here, we show that the deficiency of functional U5 small nuclear ribonucleoprotein particles (snRNPs) in Drosophila imaginal cells causes extensive transcriptome remodeling and accumulation of highly mutagenic R-loops, triggering a robust stress response and cell cycle arrest. Despite compromised proliferative capacity, the U5 snRNP-deficient cells increased protein translation and cell size, causing intra-organ growth disbalance before being gradually eliminated via apoptosis. We identify the Xrp1-Irbp18 heterodimer as the primary driver of transcriptional and cellular stress program downstream of U5 snRNP malfunction. Knockdown of Xrp1 or Irbp18 in U5 snRNP-deficient cells attenuated JNK and p53 activity, restored normal cell cycle progression and growth, and inhibited cell death. Reducing Xrp1-Irbp18, however, did not rescue the splicing defects, highlighting the requirement of accurate splicing for cellular and tissue homeostasis. Our work provides novel insights into the crosstalk between splicing and the DNA damage response and defines the Xrp1-Irbp18 heterodimer as a critical sensor of spliceosome malfunction and mediator of the stress-induced cellular senescence program.


The removal of introns and the joining of exons into mature mRNA by the spliceosome is crucial in regulating gene expression, simultaneously safeguarding genome integrity and enhancing proteome diversity in multicellular organisms. Spliceosome dysfunction is thus associated with various diseases and organismal aging. Our study describes the cascade of events in response to spliceosome dysfunction. We identified two transcription factors as drivers of a stress response program triggered by spliceosome dysfunction, which dramatically remodel gene expression to protect tissue integrity and induce a senescent-like state in damaged cells prior to their inevitable elimination. Together, we highlight the indispensable role of spliceosomes in maintaining homeostasis and implicate spliceosome dysfunction in senescent cell accumulation associated with the pathomechanisms of spliceopathies and aging.


Asunto(s)
Proteínas de Unión al ADN , Ribonucleoproteína Nuclear Pequeña U5 , Empalmosomas , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN/genética , Empalmosomas/genética , Empalmosomas/metabolismo , Animales , Drosophila melanogaster , Proteínas de Unión al ADN/metabolismo
2.
Nucleic Acids Res ; 49(3): 1688-1707, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33444449

RESUMEN

Pre-mRNA splicing catalyzed by the spliceosome represents a critical step in the regulation of gene expression contributing to transcriptome and proteome diversity. The spliceosome consists of five small nuclear ribonucleoprotein particles (snRNPs), the biogenesis of which remains only partially understood. Here we define the evolutionarily conserved protein Ecdysoneless (Ecd) as a critical regulator of U5 snRNP assembly and Prp8 stability. Combining Drosophila genetics with proteomic approaches, we demonstrate the Ecd requirement for the maintenance of adult healthspan and lifespan and identify the Sm ring protein SmD3 as a novel interaction partner of Ecd. We show that the predominant task of Ecd is to deliver Prp8 to the emerging U5 snRNPs in the cytoplasm. Ecd deficiency, on the other hand, leads to reduced Prp8 protein levels and compromised U5 snRNP biogenesis, causing loss of splicing fidelity and transcriptome integrity. Based on our findings, we propose that Ecd chaperones Prp8 to the forming U5 snRNP allowing completion of the cytoplasmic part of the U5 snRNP biogenesis pathway necessary to meet the cellular demand for functional spliceosomes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Factores de Empalme de ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U5/metabolismo , Animales , Línea Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mutación , Estabilidad Proteica , Empalme del ARN , Transcriptoma
3.
PLoS Genet ; 14(3): e1007241, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29494583

RESUMEN

Interplay between apicobasal cell polarity modules and the cytoskeleton is critical for differentiation and integrity of epithelia. However, this coordination is poorly understood at the level of gene regulation by transcription factors. Here, we establish the Drosophila activating transcription factor 3 (atf3) as a cell polarity response gene acting downstream of the membrane-associated Scribble polarity complex. Loss of the tumor suppressors Scribble or Dlg1 induces atf3 expression via aPKC but independent of Jun-N-terminal kinase (JNK) signaling. Strikingly, removal of Atf3 from Dlg1 deficient cells restores polarized cytoarchitecture, levels and distribution of endosomal trafficking machinery, and differentiation. Conversely, excess Atf3 alters microtubule network, vesicular trafficking and the partition of polarity proteins along the apicobasal axis. Genomic and genetic approaches implicate Atf3 as a regulator of cytoskeleton organization and function, and identify Lamin C as one of its bona fide target genes. By affecting structural features and cell morphology, Atf3 functions in a manner distinct from other transcription factors operating downstream of disrupted cell polarity.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Polaridad Celular/fisiología , Proteínas de Drosophila/metabolismo , Factor de Transcripción Activador 3/genética , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Inmunoprecipitación de Cromatina , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Endosomas/metabolismo , Ojo/crecimiento & desarrollo , Discos Imaginales/citología , Discos Imaginales/fisiología , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Larva , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana , Motivos de Nucleótidos/fisiología , Proteína Quinasa C/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
4.
Hum Mol Genet ; 27(10): 1772-1784, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29528393

RESUMEN

Bicaudal D2 (BICD2) encodes a highly conserved motor adaptor protein that regulates the dynein-dynactin complex in different cellular processes. Heterozygous mutations in BICD2 cause autosomal dominant lower extremity-predominant spinal muscular atrophy-2 (SMALED2). Although, various BICD2 mutations have been shown to alter interactions with different binding partners or the integrity of the Golgi apparatus, the specific pathological effects of BICD2 mutations underlying SMALED2 remain elusive. Here, we show that the fibroblasts derived from individuals with SMALED2 exhibit stable microtubules. Importantly, this effect was observed regardless of where the BICD2 mutation is located, which unifies the most likely cellular mechanism affecting microtubules. Significantly, overexpression of SMALED2-causing BICD2 mutations in the disease-relevant cell type, motor neurons, also results in an increased microtubule stability which is accompanied by axonal aberrations such as collateral branching and overgrowth. To study the pathological consequences of BICD2 mutations in vivo, and to address the controversial debate whether two of these mutations are neuron or muscle specific, we generated the first Drosophila model of SMALED2. Strikingly, neuron-specific expression of BICD2 mutants resulted in reduced neuromuscular junction size in larvae and impaired locomotion of adult flies. In contrast, expressing BICD2 mutations in muscles had no obvious effect on motor function, supporting a primarily neurological etiology of the disease. Thus, our findings contribute to the better understanding of SMALED2 pathology by providing evidence for a common pathomechanism of BICD2 mutations that increase microtubule stability in motor neurons leading to increased axonal branching and to impaired neuromuscular junction development.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas Asociadas a Microtúbulos/genética , Unión Neuromuscular/genética , Atrofias Musculares Espinales de la Infancia/genética , Animales , Modelos Animales de Enfermedad , Drosophila , Complejo Dinactina/genética , Dineínas/genética , Aparato de Golgi/genética , Aparato de Golgi/patología , Humanos , Microtúbulos/genética , Microtúbulos/patología , Mutación , Mutación Missense/genética , Unión Neuromuscular/patología , Linaje , Unión Proteica , Atrofias Musculares Espinales de la Infancia/fisiopatología
5.
PLoS Genet ; 11(7): e1005394, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26161662

RESUMEN

Juvenile hormones (JHs) play a major role in controlling development and reproduction in insects and other arthropods. Synthetic JH-mimicking compounds such as methoprene are employed as potent insecticides against significant agricultural, household and disease vector pests. However, a receptor mediating effects of JH and its insecticidal mimics has long been the subject of controversy. The bHLH-PAS protein Methoprene-tolerant (Met), along with its Drosophila melanogaster paralog germ cell-expressed (Gce), has emerged as a prime JH receptor candidate, but critical evidence that this protein must bind JH to fulfill its role in normal insect development has been missing. Here, we show that Gce binds a native D. melanogaster JH, its precursor methyl farnesoate, and some synthetic JH mimics. Conditional on this ligand binding, Gce mediates JH-dependent gene expression and the hormone's vital role during development of the fly. Any one of three different single amino acid mutations in the ligand-binding pocket that prevent binding of JH to the protein block these functions. Only transgenic Gce capable of binding JH can restore sensitivity to JH mimics in D. melanogaster Met-null mutants and rescue viability in flies lacking both Gce and Met that would otherwise die at pupation. Similarly, the absence of Gce and Met can be compensated by expression of wild-type but not mutated transgenic D. melanogaster Met protein. This genetic evidence definitively establishes Gce/Met in a JH receptor role, thus resolving a long-standing question in arthropod biology.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Hormonas Juveniles/metabolismo , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Línea Celular , Drosophila melanogaster/genética , Ácidos Grasos Insaturados/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Unión Proteica/fisiología , Transducción de Señal/genética
6.
PLoS Genet ; 10(4): e1004287, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24722212

RESUMEN

The steroid hormone ecdysone coordinates insect growth and development, directing the major postembryonic transition of forms, metamorphosis. The steroid-deficient ecdysoneless1 (ecd1) strain of Drosophila melanogaster has long served to assess the impact of ecdysone on gene regulation, morphogenesis, or reproduction. However, ecd also exerts cell-autonomous effects independently of the hormone, and mammalian Ecd homologs have been implicated in cell cycle regulation and cancer. Why the Drosophila ecd1 mutants lack ecdysone has not been resolved. Here, we show that in Drosophila cells, Ecd directly interacts with core components of the U5 snRNP spliceosomal complex, including the conserved Prp8 protein. In accord with a function in pre-mRNA splicing, Ecd and Prp8 are cell-autonomously required for survival of proliferating cells within the larval imaginal discs. In the steroidogenic prothoracic gland, loss of Ecd or Prp8 prevents splicing of a large intron from CYP307A2/spookier (spok) pre-mRNA, thus eliminating this essential ecdysone-biosynthetic enzyme and blocking the entry to metamorphosis. Human Ecd (hEcd) can substitute for its missing fly ortholog. When expressed in the Ecd-deficient prothoracic gland, hEcd re-establishes spok pre-mRNA splicing and protein expression, restoring ecdysone synthesis and normal development. Our work identifies Ecd as a novel pre-mRNA splicing factor whose function has been conserved in its human counterpart. Whether the role of mammalian Ecd in cancer involves pre-mRNA splicing remains to be discovered.


Asunto(s)
Proteínas de Drosophila/genética , Precursores del ARN/genética , Empalme del ARN/genética , Esteroides/metabolismo , Animales , Ciclo Celular/genética , Células Cultivadas , Drosophila melanogaster/genética , Ecdisona/genética , Regulación del Desarrollo de la Expresión Génica/genética , Larva/genética , Mutación/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Empalmosomas/genética
7.
J Cell Sci ; 126(Pt 4): 927-38, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23239028

RESUMEN

Cell shape dynamics, motility, and cell proliferation all depend on the actin cytoskeleton. Malignant cancer cells hijack the actin network to grow and migrate to secondary sites. Understanding the function of actin regulators is therefore of major interest. In the present study, we identify the actin cross-linking protein Filamin/Cheerio (Cher) as a mediator of malignancy in genetically defined Drosophila tumors. We show that in invasive tumors, resulting from cooperation of activated Ras with disrupted epithelial cell polarity, Cher is upregulated in a Jun N-terminal kinase (JNK)-dependent manner. Although dispensable in normal epithelium, Cher becomes required in the tumor cells for their growth and invasiveness. When deprived of Cher, these tumor clones lose their full potential to proliferate and breach tissue boundaries. Instead, the Cher-deficient clones remain confined within the limits of their source epithelium, permitting survival of the host animal. Through interaction with the myosin II heavy chain subunit, Cher is likely to strengthen the cortical actomyosin network and reinforce mechanical tension within the invasive tumors. Accordingly, Cher is required for aberrant expression of genes downstream of the Hippo/Yorkie signaling in the tumor tissue. Our study identifies Cher as a new target of JNK signaling that links cytoskeleton dynamics to tumor progression.


Asunto(s)
Proteínas Contráctiles/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias/metabolismo , Transducción de Señal/fisiología , Actinas , Animales , Línea Celular , Proteínas Contráctiles/genética , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Filaminas , Citometría de Flujo , Inmunoprecipitación , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas de Microfilamentos/genética , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Neoplasias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
8.
Development ; 137(1): 141-50, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20023169

RESUMEN

Epithelial sheet spreading and fusion underlie important developmental processes. Well-characterized examples of such epithelial morphogenetic events have been provided by studies in Drosophila, and include embryonic dorsal closure, formation of the adult thorax and wound healing. All of these processes require the basic region-leucine zipper (bZIP) transcription factors Jun and Fos. Much less is known about morphogenesis of the fly abdomen, which involves replacement of larval epidermal cells (LECs) with adult histoblasts that divide, migrate and finally fuse to form the adult epidermis during metamorphosis. Here, we implicate Drosophila Activating transcription factor 3 (Atf3), the single ortholog of human ATF3 and JDP2 bZIP proteins, in abdominal morphogenesis. During the process of the epithelial cell replacement, transcription of the atf3 gene declines. When this downregulation is experimentally prevented, the affected LECs accumulate cell-adhesion proteins and their extrusion and replacement with histoblasts are blocked. The abnormally adhering LECs consequently obstruct the closure of the adult abdominal epithelium. This closure defect can be either mimicked and further enhanced by knockdown of the small GTPase Rho1 or, conversely, alleviated by stimulating ecdysone steroid hormone signaling. Both Rho and ecdysone pathways have been previously identified as effectors of the LEC replacement. To elicit the gain-of-function effect, Atf3 specifically requires its binding partner Jun. Our data thus identify Atf3 as a new functional partner of Drosophila Jun during development.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción Activador 3/genética , Animales , Proteínas de Drosophila/genética , Ensayo de Cambio de Movilidad Electroforética , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunoprecipitación , Microscopía Confocal , Unión Proteica , Proteínas Proto-Oncogénicas c-jun/genética
9.
Life Sci Alliance ; 6(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36446522

RESUMEN

Post-transcriptional gene silencing using double-stranded RNA has revolutionized the field of functional genetics, allowing fast and easy disruption of gene function in various organisms. In Drosophila, many transgenic RNAi lines have been generated in large-scale efforts, including the Drosophila Transgenic RNAi Project (TRiP), to facilitate in vivo knockdown of virtually any Drosophila gene with spatial and temporal resolution. The available transgenic RNAi lines represent a fundamental resource for the fly community, providing an unprecedented opportunity to address a vast range of biological questions relevant to basic and biomedical research fields. However, caution should be applied regarding the efficiency and specificity of the RNAi approach. Here, we demonstrate that pVALIUM10-based RNAi lines, representing ∼13% of the total TRiP collection (1,808 of 13,410 pVALIUM TRiP-based RNAi lines), cause unintended off-target silencing of transgenes expressed from Gateway destination vectors. The silencing is mediated by targeting attB1 and attB2 sequences generated via site-specific recombination and included in the transcribed mRNA. Deleting these attB sites from the Gateway expression vector prevents silencing and restores expected transgene expression.


Asunto(s)
Drosophila , ARN Bicatenario , Animales , Interferencia de ARN , Drosophila/genética , Animales Modificados Genéticamente , Transgenes/genética , ARN Bicatenario/genética
10.
Elife ; 122023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37133250

RESUMEN

Wound response programs are often activated during neoplastic growth in tumors. In both wound repair and tumor growth, cells respond to acute stress and balance the activation of multiple programs, including apoptosis, proliferation, and cell migration. Central to those responses are the activation of the JNK/MAPK and JAK/STAT signaling pathways. Yet, to what extent these signaling cascades interact at the cis-regulatory level and how they orchestrate different regulatory and phenotypic responses is still unclear. Here, we aim to characterize the regulatory states that emerge and cooperate in the wound response, using the Drosophila melanogaster wing disc as a model system, and compare these with cancer cell states induced by rasV12scrib-/- in the eye disc. We used single-cell multiome profiling to derive enhancer gene regulatory networks (eGRNs) by integrating chromatin accessibility and gene expression signals. We identify a 'proliferative' eGRN, active in the majority of wounded cells and controlled by AP-1 and STAT. In a smaller, but distinct population of wound cells, a 'senescent' eGRN is activated and driven by C/EBP-like transcription factors (Irbp18, Xrp1, Slow border, and Vrille) and Scalloped. These two eGRN signatures are found to be active in tumor cells at both gene expression and chromatin accessibility levels. Our single-cell multiome and eGRNs resource offers an in-depth characterization of the senescence markers, together with a new perspective on the shared gene regulatory programs acting during wound response and oncogenesis.


Asunto(s)
Proteínas de Drosophila , Neoplasias , Animales , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Redes Reguladoras de Genes , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias/patología , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo
11.
FEBS J ; 289(15): 4497-4517, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35191183

RESUMEN

Cancer development has been linked to aberrant sensing and interpretation of mechanical cues and force-generating properties. Here, we show that upregulation of the actin crosslinking protein Cheerio (Cher), the fly ortholog of Filamin A (FLNA), and the conformation of its mechanosensitive region (MSR) are instrumental to the malignancy of polarity-deficient, Ras-driven tumours in Drosophila epithelia. We demonstrate that impaired growth and cytoskeletal contractility of tumours devoid of cher can be rescued by stimulating myosin activity. Profiling the Cher interactome in tumour-bearing imaginal discs identified several components of the cell cortex, including the ß-heavy Spectrin Karst (Kst), the scaffolding protein Big bang (Bbg), and 14-3-3ε. We show that Cher binds Bbg through the MSR while the interaction with 14-3-3ε and Kst is MSR-independent. Importantly, our genetic studies define Bbg, Kst, and 14-3-3ε as tumour suppressors. The tumour-promoting function of Cher thus relies on its capacity to control the contractile state of the cytoskeleton through interactions with myosin and specific components of the cell cortex.


Asunto(s)
Actinas , Neoplasias , Actinas/metabolismo , Animales , Carcinogénesis/genética , Drosophila/genética , Filaminas/genética , Filaminas/metabolismo
12.
Elife ; 92020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33026342

RESUMEN

Blood development in multicellular organisms relies on specific tissue microenvironments that nurture hematopoietic precursors and promote their self-renewal, proliferation, and differentiation. The mechanisms driving blood cell homing and their interactions with hematopoietic microenvironments remain poorly understood. Here, we use the Drosophila melanogaster model to reveal a pivotal role for basement membrane composition in the formation of hematopoietic compartments. We demonstrate that by modulating extracellular matrix components, the fly blood cells known as hemocytes can be relocated to tissue surfaces where they function similarly to their natural hematopoietic environment. We establish that the Collagen XV/XVIII ortholog Multiplexin in the tissue-basement membranes and the phagocytosis receptor Eater on the hemocytes physically interact and are necessary and sufficient to induce immune cell-tissue association. These results highlight the cooperation of Multiplexin and Eater as an integral part of a homing mechanism that specifies and maintains hematopoietic sites in Drosophila.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/genética , Colágeno/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Hematopoyesis/genética , Sistema Hematopoyético/metabolismo , Receptores de Superficie Celular/genética , Animales , Membrana Basal/metabolismo , Diferenciación Celular , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Colágeno/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Receptores de Superficie Celular/metabolismo
13.
Dis Model Mech ; 13(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32424050

RESUMEN

Retinitis pigmentosa (RP) represents genetically heterogeneous and clinically variable disease characterized by progressive degeneration of photoreceptors resulting in a gradual loss of vision. The autosomal dominant RP type 13 (RP13) has been linked to the malfunction of PRPF8, an essential component of the spliceosome. Over 20 different RP-associated PRPF8 mutations have been identified in human patients. However, the cellular and molecular consequences of their expression in vivo in specific tissue contexts remain largely unknown. Here, we establish a Drosophila melanogaster model for RP13 by introducing the nine distinct RP mutations into the fly PRPF8 ortholog prp8 and express the mutant proteins in precise spatiotemporal patterns using the Gal4/UAS system. We show that all nine RP-Prp8 mutant proteins negatively impact developmental timing, albeit to a different extent, when expressed in the endocrine cells producing the primary insect moulting hormone. In the developing eye primordium, uncommitted epithelial precursors rather than differentiated photoreceptors appeared sensitive to Prp8 malfunction. Expression of the two most pathogenic variants, Prp8S>F and Prp8H>R, induced apoptosis causing alterations to the adult eye morphology. The affected tissue mounted stress and cytoprotective responses, while genetic programs underlying neuronal function were attenuated. Importantly, the penetrance and expressivity increased under prp8 heterozygosity. In contrast, blocking apoptosis alleviated cell loss but not the redox imbalance. Remarkably, the pathogenicity of the RP-Prp8 mutations in Drosophila correlates with the severity of clinical phenotypes in patients carrying the equivalent mutations, highlighting the suitability of the Drosophila model for in-depth functional studies of the mechanisms underlying RP13 etiology.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Mutación , Células Fotorreceptoras de Vertebrados/patología , Factores de Empalme de ARN/genética , Retinitis Pigmentosa/genética , Animales , Animales Modificados Genéticamente , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Heterocigoto , Masculino , Morfogénesis , Fenotipo , Células Fotorreceptoras de Vertebrados/metabolismo , Factores de Empalme de ARN/metabolismo , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
14.
Elife ; 82019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30735120

RESUMEN

The restoration of homeostasis after tissue damage relies on proper spatial-temporal control of damage-induced apoptosis and compensatory proliferation. In Drosophila imaginal discs these processes are coordinated by the stress response pathway JNK. We demonstrate that JNK signaling induces a dose-dependent extension of G2 in tissue damage and tumors, resulting in either transient stalling or a prolonged but reversible cell cycle arrest. G2-stalling is mediated by downregulation of the G2/M-specific phosphatase String(Stg)/Cdc25. Ectopic expression of stg is sufficient to suppress G2-stalling and reveals roles for stalling in survival, proliferation and paracrine signaling. G2-stalling protects cells from JNK-induced apoptosis, but under chronic conditions, reduces proliferative potential of JNK-signaling cells while promoting non-autonomous proliferation. Thus, transient cell cycle stalling in G2 has key roles in wound healing but becomes detrimental upon chronic JNK overstimulation, with important implications for chronic wound healing pathologies or tumorigenic transformation.


Asunto(s)
Senescencia Celular/genética , Discos Imaginales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Estrés Fisiológico/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Puntos de Control del Ciclo Celular/genética , División Celular/genética , Proliferación Celular/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Humanos , Discos Imaginales/crecimiento & desarrollo , Discos Imaginales/lesiones , Sistema de Señalización de MAP Quinasas/genética , Cicatrización de Heridas/genética
15.
Cell Rep ; 27(10): 3019-3033.e5, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167145

RESUMEN

Homeostatic renewal and stress-related tissue regeneration rely on stem cell activity, which drives the replacement of damaged cells to maintain tissue integrity and function. The Jun N-terminal kinase (JNK) signaling pathway has been established as a critical regulator of tissue homeostasis both in intestinal stem cells (ISCs) and mature enterocytes (ECs), while its chronic activation has been linked to tissue degeneration and aging. Here, we show that JNK signaling requires the stress-inducible transcription factor Ets21c to promote tissue renewal in Drosophila. We demonstrate that Ets21c controls ISC proliferation as well as EC apoptosis through distinct sets of target genes that orchestrate cellular behaviors via intrinsic and non-autonomous signaling mechanisms. While its loss appears dispensable for development and prevents epithelial aging, ISCs and ECs demand Ets21c function to mount cellular responses to oxidative stress. Ets21c thus emerges as a vital regulator of proliferative homeostasis in the midgut and a determinant of the adult healthspan.


Asunto(s)
Envejecimiento , Proteínas de Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Apoptosis , Proliferación Celular , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas del Huevo/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Mucosa Intestinal/citología , Longevidad , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Unión Proteica , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo
16.
J Vis Exp ; (116)2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27768082

RESUMEN

Drosophila melanogaster has emerged as a powerful experimental system for functional and mechanistic studies of tumor development and progression in the context of a whole organism. Sophisticated techniques to generate genetic mosaics facilitate induction of visually marked, genetically defined clones surrounded by normal tissue. The clones can be analyzed through diverse molecular, cellular and omics approaches. This study describes how to generate fluorescently labeled clonal tumors of varying malignancy in the eye/antennal imaginal discs (EAD) of Drosophila larvae using the Mosaic Analysis with a Repressible Cell Marker (MARCM) technique. It describes procedures how to recover the mosaic EAD and brain from the larvae and how to process them for simultaneous imaging of fluorescent transgenic reporters and antibody staining. To facilitate molecular characterization of the mosaic tissue, we describe a protocol for isolation of total RNA from the EAD. The dissection procedure is suitable to recover EAD and brains from any larval stage. The fixation and staining protocol for imaginal discs works with a number of transgenic reporters and antibodies that recognize Drosophila proteins. The protocol for RNA isolation can be applied to various larval organs, whole larvae, and adult flies. Total RNA can be used for profiling of gene expression changes using candidate or genome-wide approaches. Finally, we detail a method for quantifying invasiveness of the clonal tumors. Although this method has limited use, its underlying concept is broadly applicable to other quantitative studies where cognitive bias must be avoided.


Asunto(s)
Drosophila melanogaster , Regulación del Desarrollo de la Expresión Génica , Discos Imaginales , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila , Larva , Microscopía Confocal , Mosaicismo , Neoplasias
17.
Dis Model Mech ; 8(10): 1279-93, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26398940

RESUMEN

Cancer initiation and maintenance of the transformed cell state depend on altered cellular signaling and aberrant activities of transcription factors (TFs) that drive pathological gene expression in response to cooperating genetic lesions. Deciphering the roles of interacting TFs is therefore central to understanding carcinogenesis and for designing cancer therapies. Here, we use an unbiased genomic approach to define a TF network that triggers an abnormal gene expression program promoting malignancy of clonal tumors, generated in Drosophila imaginal disc epithelium by gain of oncogenic Ras (Ras(V12)) and loss of the tumor suppressor Scribble (scrib(1)). We show that malignant transformation of the ras(V12)scrib(1) tumors requires TFs of distinct families, namely the bZIP protein Fos, the ETS-domain factor Ets21c and the nuclear receptor Ftz-F1, all acting downstream of Jun-N-terminal kinase (JNK). Depleting any of the three TFs improves viability of tumor-bearing larvae, and this positive effect can be enhanced further by their combined removal. Although both Fos and Ftz-F1 synergistically contribute to ras(V12)scrib(1) tumor invasiveness, only Fos is required for JNK-induced differentiation defects and Matrix metalloprotease (MMP1) upregulation. In contrast, the Fos-dimerizing partner Jun is dispensable for JNK to exert its effects in ras(V12)scrib(1) tumors. Interestingly, Ets21c and Ftz-F1 are transcriptionally induced in these tumors in a JNK- and Fos-dependent manner, thereby demonstrating a hierarchy within the tripartite TF network, with Fos acting as the most upstream JNK effector. Of the three TFs, only Ets21c can efficiently substitute for loss of polarity and cooperate with Ras(V12) in inducing malignant clones that, like ras(V12)scrib(1) tumors, invade other tissues and overexpress MMP1 and the Drosophila insulin-like peptide 8 (Dilp8). While ras(V12)ets21c tumors require JNK for invasiveness, the JNK activity is dispensable for their growth. In conclusion, our study delineates both unique and overlapping functions of distinct TFs that cooperatively promote aberrant expression of target genes, leading to malignant tumor phenotypes.


Asunto(s)
Carcinogénesis/patología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinogénesis/genética , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Modelos Biológicos , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Oncogenes , ARN Mensajero/genética , ARN Mensajero/metabolismo , Supresión Genética , Transcriptoma/genética , Proteínas ras/metabolismo
18.
Mol Cell Biol ; 32(19): 3949-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22851689

RESUMEN

Integration of metabolic and immune responses during animal development ensures energy balance, permitting both growth and defense. Disturbed homeostasis causes organ failure, growth retardation, and metabolic disorders. Here, we show that the Drosophila melanogaster activating transcription factor 3 (Atf3) safeguards metabolic and immune system homeostasis. Loss of Atf3 results in chronic inflammation and starvation responses mounted primarily by the larval gut epithelium, while the fat body suffers lipid overload, causing energy imbalance and death. Hyperactive proinflammatory and stress signaling through NF-κB/Relish, Jun N-terminal kinase, and FOXO in atf3 mutants deregulates genes important for immune defense, digestion, and lipid metabolism. Reducing the dose of either FOXO or Relish normalizes both lipid metabolism and gene expression in atf3 mutants. The function of Atf3 is conserved, as human ATF3 averts some of the Drosophila mutant phenotypes, improving their survival. The single Drosophila Atf3 may incorporate the diversified roles of two related mammalian proteins.


Asunto(s)
Factor de Transcripción Activador 3/inmunología , Proteínas de Drosophila/inmunología , Drosophila melanogaster/fisiología , Homeostasis , Inmunidad , Metabolismo de los Lípidos , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 3/uso terapéutico , Animales , Digestión , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/microbiología , Grasas/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Mutación , Obesidad/genética , Inanición/genética
19.
Dev Cell ; 18(1): 157-64, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20152186

RESUMEN

FGF signaling is a central regulator of branching morphogenesis processes, such as angiogenesis or the development of branched organs including lung, kidney, and mammary gland. The formation of the air sac during the development of the Drosophila tracheal system is a powerful genetic model to investigate how FGF signaling patterns such emerging structures. This article describes the characterization of the Drosophila matrix metalloprotease Mmp2 as an extracellular inhibitor of FGF morphogenetic function. Mmp2 expression in the developing air sac is controlled by the Drosophila FGF homolog Branchless and then participates in a negative feedback and lateral inhibition mechanism that defines the precise pattern of FGF signaling. The signaling function for MMPs described here may not be limited to branching morphogenesis processes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Morfogénesis/fisiología , Sistema Respiratorio/embriología , Sistema Respiratorio/enzimología , Animales , Proteínas de Drosophila/genética , Retroalimentación Fisiológica/fisiología , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Metaloproteinasa 2 de la Matriz/genética , Organogénesis/fisiología , Transducción de Señal/fisiología
20.
EMBO J ; 25(22): 5294-304, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17082773

RESUMEN

Loss of the epithelial polarity gene scribble in clones of Drosophila imaginal disc cells can cooperate with Ras signaling to induce malignant tumors. Such mutant tissue overproliferates, resists apoptosis, leaves its place of origin and invades other organs, ultimately causing lethality. We show that increased Jun N-terminal kinase (JNK) signaling resulting from the loss of scribble promotes the movement of transformed cells to secondary sites. This effect requires Fos-dependent transcriptional activation of a matrix metalloprotease gene mmp1 downstream of JNK. Expression of the Mmp inhibitor Timp or Mmp RNAi knockdown suppresses cell invasiveness. The proinvasive function of the JNK pathway is revealed in a tumor context when active Ras signaling prevents the apoptotic response to JNK activity as it occurs in nontransformed cells. Based on these results, we present a model that explains the oncogenic cooperation between JNK and Ras, and describes how aberrant regulation of cell survival, proliferation and mobilization cooperate to incite malignant tumor formation.


Asunto(s)
Proteínas de Drosophila/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Metaloproteinasa 1 de la Matriz/fisiología , Invasividad Neoplásica , Proteínas ras/fisiología , Animales , Apoptosis , Movimiento Celular , Polaridad Celular , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 1 de la Matriz/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Modelos Biológicos , Mutación , Transducción de Señal , Inhibidores Tisulares de Metaloproteinasas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA