Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Int J Mol Sci ; 24(16)2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37629022

RESUMEN

Ankyrin repeat and single KH domain-containing protein 1 (ANKHD1) is a large, scaffolding protein composed of two stretches of ankyrin repeat domains that mediate protein-protein interactions and a KH domain that mediates RNA or single-stranded DNA binding. ANKHD1 interacts with proteins in several crucial signalling pathways, including receptor tyrosine kinase, JAK/STAT, mechanosensitive Hippo (YAP/TAZ), and p21. Studies into the role of ANKHD1 in cancer cell lines demonstrate a crucial role in driving uncontrolled cellular proliferation and growth, enhanced tumorigenicity, cell cycle progression through the S phase, and increased epithelial-to-mesenchymal transition. Furthermore, at a clinical level, the increased expression of ANKHD1 has been associated with greater tumour infiltration, increased metastasis, and larger tumours. Elevated ANKHD1 resulted in poorer prognosis, more aggressive growth, and a decrease in patient survival in numerous cancer types. This review aims to gather the current knowledge about ANKHD1 and explore its molecular properties and functions, focusing on the protein's role in cancer at both a cellular and clinical level.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Hiperplasia , Agresión , Repetición de Anquirina , División Celular , Proteínas de Unión al ARN
2.
J Cell Sci ; 131(13)2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29848658

RESUMEN

Cytokine receptors often act via the Janus kinase and signal transducer and activator of transcription (JAK/STAT) pathway to form a signalling cascade that is essential for processes such as haematopoiesis, immune responses and tissue homeostasis. In order to transduce ligand activation, cytokine receptors must dimerise. However, mechanisms regulating their dimerisation are poorly understood. In order to better understand the processes regulating cytokine receptor levels, and their activity and dimerisation, we analysed the highly conserved JAK/STAT pathway in Drosophila, which acts via a single receptor, known as Domeless. We performed a genome-wide RNAi screen in Drosophila cells, identifying MASK as a positive regulator of Domeless dimerisation and protein levels. We show that MASK is able to regulate receptor levels and JAK/STAT signalling both in vitro and in vivo We also show that its human homologue, ANKHD1, is also able to regulate JAK/STAT signalling and the levels of a subset of pathway receptors in human cells. Taken together, our results identify MASK as a novel regulator of cytokine receptor levels, and suggest functional conservation, which may have implications for human health.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Genoma de los Insectos , Interferencia de ARN , Receptores de Citocinas/genética , Receptores de Interleucina/química , Secuencias de Aminoácidos , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Unión Proteica , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal
3.
Biochem Soc Trans ; 48(2): 559-567, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32239204

RESUMEN

Developed over 70 years ago as an anti-folate chemotherapy agent, methotrexate (MTX) is a WHO 'essential medicine' that is now widely employed as a first-line treatment in auto-immune, inflammatory diseases such as rheumatoid arthritis (RA), psoriasis and Crone's disease. When used for these diseases patients typically take a once weekly low-dose of MTX - a therapy which provides effective inflammatory control to tens of millions of people worldwide. While undoubtedly effective, our understanding of the anti-inflammatory mechanism-of-action of low-dose MTX is incomplete. In particular, the long-held dogma that this disease-modifying anti-rheumatic drug (DMARD) acts via the folate pathway does not appear to hold up to scrutiny. Recently, MTX has been identified as an inhibitor of JAK/STAT pathway activity, a suggestion supported by many independent threads of evidence. Intriguingly, the JAK/STAT pathway is central to both the inflammatory and immune systems and is a pathway already targeted by other RA treatments. We suggest that the DMARD activity of MTX is likely to be largely mediated by its inhibition of JAK/STAT pathway signalling while many of its side effects are likely associated with the folate pathway. This insight into the mechanism-of-action of MTX opens the possibility for repurposing this low cost, safe and effective drug for the treatment of other JAK/STAT pathway-associated diseases.


Asunto(s)
Antirreumáticos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Metotrexato/farmacología , Artritis Reumatoide/tratamiento farmacológico , Línea Celular , Enfermedad de Crohn/tratamiento farmacológico , Ácido Fólico/metabolismo , Humanos , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas , Psoriasis/tratamiento farmacológico , Factores de Transcripción STAT/metabolismo , Transducción de Señal
4.
J Biol Chem ; 293(25): 9570-9579, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29695508

RESUMEN

Clear cell renal cell carcinoma (ccRCC) represents the most common kidney cancer worldwide. Increased cell proliferation associated with abnormal microRNA (miRNA) regulation are hallmarks of carcinogenesis. Ankyrin repeat and single KH domain 1 (ANKHD1) is a highly conserved protein found to interact with core cancer pathways in Drosophila; however, its involvement in RCC is completely unexplored. Quantitative PCR studies coupled with large-scale genomics data sets demonstrated that ANKHD1 is significantly up-regulated in kidneys of RCC patients when compared with healthy controls. Cell cycle analysis revealed that ANKHD1 is an essential factor for RCC cell division. To understand the molecular mechanism(s) utilized by ANKHD1 to drive proliferation, we performed bioinformatics analyses that revealed that ANKHD1 contains a putative miRNA-binding motif. We screened 48 miRNAs with tumor-enhancing or -suppressing activities and found that ANKHD1 binds to and regulates three tumor-suppressing miRNAs (i.e. miR-29a, miR-205, and miR-196a). RNA-immunoprecipitation assays demonstrated that ANKHD1 physically interacts with its target miRNAs via a single K-homology domain, located in the C terminus of the protein. Functionally, we discovered that ANKHD1 positively drives ccRCC cell mitosis via binding to and suppressing mainly miR-29a and to a lesser degree via miR-196a/205, leading to up-regulation in proliferative genes such as CCDN1. Collectively, these data identify ANKHD1 as a new regulator of ccRCC proliferation via specific miRNA interactions.


Asunto(s)
Carcinoma de Células Renales/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/patología , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Sitios de Unión , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Perfilación de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Pronóstico , Proteínas de Unión al ARN/genética , Células Tumorales Cultivadas
5.
J Cell Sci ; 127(Pt 1): 101-10, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24163435

RESUMEN

JAK/STAT signalling regulates many essential developmental processes including cell proliferation and haematopoiesis, whereas its inappropriate activation is associated with the majority of myeloproliferative neoplasias and numerous cancers. Furthermore, high levels of JAK/STAT pathway signalling have also been associated with enhanced metastatic invasion by cancerous cells. Strikingly, gain-of-function mutations in the single Drosophila JAK homologue, Hopscotch, result in haemocyte neoplasia, inappropriate differentiation and the formation of melanised haemocyte-derived 'tumour' masses; phenotypes that are partly orthologous to human gain-of-function JAK2-associated pathologies. Here we show that Gα73B, a novel JAK/STAT pathway target gene, is necessary for JAK/STAT-mediated tumour formation in flies. In addition, although Gα73B does not affect haemocyte differentiation, it does regulate haemocyte morphology and motility under non-pathological conditions. We show that Gα73B is required for constitutive, but not injury-induced, activation of Rho1 and for the localisation of Rho1 into filopodia upon haemocyte activation. Consistent with these results, we also show that Rho1 interacts genetically with JAK/STAT signalling, and that wild-type levels of Rho1 are necessary for tumour formation. Our findings link JAK/STAT transcriptional outputs, Gα73B activity and Rho1-dependent cytoskeletal rearrangements and cell motility, therefore connecting a pathway associated with cancer with a marker indicative of invasiveness. As such, we suggest a mechanism by which JAK/STAT pathway signalling may promote metastasis.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Regulación Neoplásica de la Expresión Génica , Hematopoyesis/genética , Hemocitos/metabolismo , Quinasas Janus/genética , Factores de Transcripción STAT/genética , Factores de Transcripción/genética , Proteínas de Unión al GTP rho/genética , Animales , Movimiento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hemocitos/patología , Quinasas Janus/metabolismo , Masculino , Seudópodos/metabolismo , Seudópodos/patología , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP rho/metabolismo
6.
Development ; 140(18): 3858-68, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23946443

RESUMEN

Morphogenesis is dependent on the orchestration of multiple developmental processes to generate mature functional organs. However, the signalling pathways that coordinate morphogenesis and the mechanisms that translate these signals into tissue shape changes are not well understood. Here, we demonstrate that changes in intercellular adhesion mediated by the transmembrane protein Fasciclin III (FasIII) represent a key mediator of morphogenesis. Using the embryonic Drosophila hindgut as an in vivo model for organogenesis, we show that the tightening of hindgut curvature that normally occurs between embryonic stage 12 and 15 to generate the characteristic shepherd's crook shape is dependent on localised JAK/STAT pathway activation. This localised pathway activity drives the expression of FasIII leading to its subcellular lateralisation at a stage before formation of septate junctions. Additionally, we show that JAK/STAT- and FasIII-dependent morphogenesis also regulates folds within the third instar wing imaginal disc. We show that FasIII forms homophilic intercellular interactions that promote intercellular adhesion in vivo and in cultured cells. To explore these findings, we have developed a mathematical model of the developing hindgut, based on the differential interfacial tension hypothesis (DITH) linking intercellular adhesion and localised surface tension. Our model suggests that increased intercellular adhesion provided by FasIII can be sufficient to drive the tightening of tube curvature observed. Taken together, these results identify a conserved molecular mechanism that directly links JAK/STAT pathway signalling to intercellular adhesion and that sculpts both tubular and planar epithelial shape.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/citología , Tracto Gastrointestinal/anatomía & histología , Tracto Gastrointestinal/citología , Animales , Adhesión Celular , Drosophila melanogaster/embriología , Drosophila melanogaster/enzimología , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/metabolismo , Quinasas Janus/metabolismo , Modelos Biológicos , Transporte de Proteínas , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Fracciones Subcelulares/metabolismo , Alas de Animales/anatomía & histología , Alas de Animales/metabolismo
8.
Front Med (Lausanne) ; 11: 1285772, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698784

RESUMEN

JAK/STAT pathway signalling is associated with both chronic inflammatory conditions such as psoriasis and haematological malignancies such as the myeloproliferative neoplasms (MPNs). Here we describe a 73yo female patient with a history of chronic plaque psoriasis, post-essential thrombocythemia myelofibrosis (MF) and a quality of life substantially impacted by both conditions. We report that 15 mg oral Methotrexate (MTX) weekly as a monotherapy is well tolerated, provides a substantial clinical improvement for both conditions and significantly improves quality of life. We suggest that the recently identified mechanism of action of MTX as a JAK inhibitor is likely to explain this efficacy and suggest that repurposing MTX for MPNs may represent a clinical- and cost-effective therapeutic option.

9.
Front Immunol ; 14: 1310117, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38283366

RESUMEN

In Drosophila blood, plasmatocytes of the haemocyte lineage represent the functional equivalent of vertebrate macrophages and have become an established in vivo model with which to study macrophage function and behaviour. However, the use of plasmatocytes as a macrophage model has been limited by a historical perspective that plasmatocytes represent a homogenous population of cells, in contrast to the high levels of heterogeneity of vertebrate macrophages. Recently, a number of groups have reported transcriptomic approaches which suggest the existence of plasmatocyte heterogeneity, while we identified enhancer elements that identify subpopulations of plasmatocytes which exhibit potentially pro-inflammatory behaviours, suggesting conservation of plasmatocyte heterogeneity in Drosophila. These plasmatocyte subpopulations exhibit enhanced responses to wounds and decreased rates of efferocytosis when compared to the overall plasmatocyte population. Interestingly, increasing the phagocytic requirement placed upon plasmatocytes is sufficient to decrease the size of these plasmatocyte subpopulations in the embryo. However, the mechanistic basis for this response was unclear. Here, we examine how plasmatocyte subpopulations are modulated by apoptotic cell clearance (efferocytosis) demands and associated signalling pathways. We show that loss of the phosphatidylserine receptor Simu prevents an increased phagocytic burden from modulating specific subpopulation cells, while blocking other apoptotic cell receptors revealed no such rescue. This suggests that Simu-dependent efferocytosis is specifically involved in determining fate of particular subpopulations. Supportive of our original finding, mutations in amo (the Drosophila homolog of PKD2), a calcium-permeable channel which operates downstream of Simu, phenocopy simu mutants. Furthermore, we show that Amo is involved in the acidification of the apoptotic cell-containing phagosomes, suggesting that this reduction in pH may be associated with macrophage reprogramming. Additionally, our results also identify Ecdysone receptor signalling, a pathway related to control of cell death during developmental transitions, as a controller of plasmatocyte subpopulation identity. Overall, these results identify fundamental pathways involved in the specification of plasmatocyte subpopulations and so further validate Drosophila plasmatocytes as a heterogeneous population of macrophage-like cells within this important developmental and immune model.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Drosophila melanogaster/genética , Eferocitosis , Macrófagos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
10.
BMC Genomics ; 13: 506, 2012 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-23006893

RESUMEN

BACKGROUND: Genome-scale RNA-interference (RNAi) screens are becoming ever more common gene discovery tools. However, whilst every screen identifies interacting genes, less attention has been given to how factors such as library design and post-screening bioinformatics may be effecting the data generated. RESULTS: Here we present a new genome-wide RNAi screen of the Drosophila JAK/STAT signalling pathway undertaken in the Sheffield RNAi Screening Facility (SRSF). This screen was carried out using a second-generation, computationally optimised dsRNA library and analysed using current methods and bioinformatic tools. To examine advances in RNAi screening technology, we compare this screen to a biologically very similar screen undertaken in 2005 with a first-generation library. Both screens used the same cell line, reporters and experimental design, with the SRSF screen identifying 42 putative regulators of JAK/STAT signalling, 22 of which verified in a secondary screen and 16 verified with an independent probe design. Following reanalysis of the original screen data, comparisons of the two gene lists allows us to make estimates of false discovery rates in the SRSF data and to conduct an assessment of off-target effects (OTEs) associated with both libraries. We discuss the differences and similarities between the resulting data sets and examine the relative improvements in gene discovery protocols. CONCLUSIONS: Our work represents one of the first direct comparisons between first- and second-generation libraries and shows that modern library designs together with methodological advances have had a significant influence on genome-scale RNAi screens.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Genoma de los Insectos , Quinasas Janus/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción STAT/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Biblioteca de Genes , Genes Reporteros , Estudio de Asociación del Genoma Completo , Quinasas Janus/metabolismo , Luciferasas , Plásmidos , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Transfección
11.
J Cell Sci ; 123(Pt 20): 3457-66, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20841381

RESUMEN

Appropriate regulation of signal transduction pathways is essential for normal development and is often disrupted in disease. Therefore, many regulatory mechanisms and feedback loops have evolved to ensure appropriate signalling. One mechanism previously suggested to modulate a range of signal transduction pathways involves the internalisation and destruction of transmembrane receptors by the endocytic trafficking machinery. Strikingly, a recent report has suggested that the endocytic trafficking of the Drosophila JAK-STAT pathway receptor Domeless (Dome) does not act to downregulate pathway activity, but rather is necessary for in vivo signalling. Here, we examine this relationship to address the interaction of Drosophila JAK-STAT pathway signalling and endocytic trafficking. We show that Dome is trafficked through clathrin-mediated endocytosis and a directed RNAi screen identified several components of the endocytic machinery as negative regulators of pathway signalling. We demonstrate that Dome signals both from the plasma membrane and internalised vesicles and show, using knockdown experiments, that endocytic components negatively regulate JAK-STAT signalling in vivo. As such, disruption in endocytic trafficking represents a potent negative regulator of the disease relevant JAK-STAT signalling cascade.


Asunto(s)
Proteínas de Drosophila/metabolismo , Endocitosis/fisiología , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Subunidades alfa de Complejo de Proteína Adaptadora/genética , Subunidades alfa de Complejo de Proteína Adaptadora/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila , Proteínas de Drosophila/genética , Endocitosis/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Quinasas Janus/genética , Reacción en Cadena de la Polimerasa , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Interferencia de ARN , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Transcripción STAT/genética , Transducción de Señal/genética , Factores de Transcripción/genética
13.
EMBO Rep ; 11(3): 201-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20168330

RESUMEN

Although many signal transduction pathways have been implicated in the development of human disease, the identification of pathway targets and the biological processes that mediate disease progression remains challenging. One such disease-related pathway is the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) cascade whose constitutive misactivation by the JAK2 V617F mutation underlies most human myeloproliferative disorders. Here, we use transcript profiling of Drosophila haemocyte-like cells to identify JAK/STAT target genes, combined with an in vivo model for JAK-induced blood cell overproliferation, to identify the main effectors required for haematopoietic tumour development. The identified human homologues of the Drosophila effectors were tested for potential V617F-mediated transcriptional regulation in human HeLa cells and compared with small interfering RNA-derived data, quantify their role in regulating the proliferation of cancer-derived cell lines. Such an inter-species approach is an effective way to identify factors with conserved functions that might be central to human disease.


Asunto(s)
Perfilación de la Expresión Génica , Neoplasias Hematológicas/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Transcripción Genética , Animales , Proliferación Celular , Análisis por Conglomerados , Drosophila melanogaster , Regulación de la Expresión Génica , Células HeLa , Neoplasias Hematológicas/patología , Hemocitos/citología , Humanos , Ratones , ARN Interferente Pequeño/metabolismo
14.
J Gerontol A Biol Sci Med Sci ; 77(8): 1494-1502, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34137822

RESUMEN

Over recent decades, increased longevity has not been paralleled by extended health span, resulting in more years spent with multiple diseases in older age. As such, interventions to improve health span are urgently required. Zoledronate (Zol) is a nitrogen-containing bisphosphonate, which inhibits the farnesyl pyrophosphate synthase enzyme, central to the mevalonate pathway. It is already used clinically to prevent fractures in osteoporotic patients, who have been reported to derive unexpected and unexplained survival benefits. Using Drosophila as a model we determined the effects of Zol on life span, parameters of health span (climbing ability and intestinal dysplasia), and the ability to confer resistance to oxidative stress using a combination of genetically manipulated Drosophila strains and Western blotting. Our study shows that Zol extended life span, improved climbing activity, and reduced intestinal epithelial dysplasia and permeability with age. Mechanistic studies showed that Zol conferred resistance to oxidative stress and reduced accumulation of X-ray-induced DNA damage via inhibition of farnesyl pyrophosphate synthase. Moreover, Zol was associated with inhibition of phosphorylated AKT in the mammalian traget of rapamycin pathway downstream of the mevalonate pathway and required dFOXO for its action, both molecules associated with increased longevity. Taken together, our work indicates that Zol, a drug already widely used to prevent osteoporosis and dosed only once a year, modulates important mechanisms of aging. Its repurposing holds great promise as a treatment to improve health span.


Asunto(s)
Proteínas de Drosophila , Ácido Mevalónico , Animales , Línea Celular Tumoral , Drosophila , Proteínas de Drosophila/metabolismo , Factores de Transcripción Forkhead , Imidazoles/farmacología , Mamíferos , Ácido Mevalónico/metabolismo , Ácido Zoledrónico/farmacología
15.
Nature ; 436(7052): 871-5, 2005 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-16094372

RESUMEN

Signalling pathways mediating the transduction of information between cells are essential for development, cellular differentiation and homeostasis. Their dysregulation is also frequently associated with human malignancies. The Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) pathway represents one such signalling cascade whose evolutionarily conserved roles include cell proliferation and haematopoiesis. Here we describe a systematic genome-wide survey for genes required for JAK/STAT pathway activity. Analysis of 20,026 RNA interference (RNAi)-induced phenotypes in cultured Drosophila melanogaster haemocyte-like cells identified interacting genes encoding 4 known and 86 previously uncharacterized proteins. Subsequently, cell-based epistasis experiments were used to classify these proteins on the basis of their interaction with known components of the signalling cascade. In addition to multiple human disease gene homologues, we have found the tyrosine phosphatase Ptp61F and the Drosophila homologue of BRWD3, a bromo-domain-containing protein disrupted in leukaemia. Moreover, in vivo analysis demonstrates that disrupted dBRWD3 and overexpressed Ptp61F function as suppressors of leukaemia-like blood cell tumours. This screen represents a comprehensive identification of novel loci required for JAK/STAT signalling and provides molecular insights into an important pathway relevant for human cancer. Human homologues of identified pathway modifiers may constitute targets for therapeutic interventions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Genómica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Interferencia de ARN , Transducción de Señal , Transactivadores/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Epistasis Genética , Genoma , Hemocitos/citología , Hemocitos/enzimología , Hemocitos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Fenotipo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras , Factor de Transcripción STAT1 , Transactivadores/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
EMBO Rep ; 9(11): 1114-20, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18802449

RESUMEN

In vertebrates, seven signal transducer and activator of transcription (STAT) proteins bind to palindromic sites separated by spacers of two or three nucleotides (STAT1), four nucleotides (STAT6) or three nucleotides (STAT2 to STAT5a/b). This diversity of binding sites provides specificity to counter semiredundancy and was thought to be a recent evolutionary acquisition. Here, we examine the natural DNA-binding sites of the single Drosophila Stat and show that this is not the case. Rather, Drosophila Stat92E is able to bind to and activate target gene expression through both 3n and 4n spaced sites. Our experiments indicate that Stat92E has a higher binding affinity for 3n sites than for 4n sites and suggest that the levels of target gene expression can be modulated by insertion and/or deletion of single bases. Our results indicate that the ancestral STAT protein had the capacity to bind to 3n and 4n sites and that specific STAT binding preferences evolved with the radiation of the vertebrate STAT family.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Secuencia de Bases , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Evolución Molecular , Datos de Secuencia Molecular , Receptores de Interleucina/genética , Factores de Transcripción STAT/química , Activación Transcripcional
17.
Curr Biol ; 16(1): 80-8, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16401426

RESUMEN

A limited number of evolutionarily conserved signal transduction pathways are repeatedly reused during development to regulate a wide range of processes. Here we describe a new negative regulator of JAK/STAT signaling and identify a potential mechanism by which the pleiotropy of responses resulting from pathway activation is generated in vivo. As part of a genetic interaction screen, we have identified Ken & Barbie (Ken) , which is an ortholog of the mammalian proto-oncogene BCL6 , as a negative regulator of the JAK/STAT pathway. Ken genetically interacts with the pathway in vivo and recognizes a DNA consensus sequence overlapping that of STAT92E in vitro. Tissue culture-based assays demonstrate the existence of Ken-sensitive and Ken-insensitive STAT92E binding sites, while ectopically expressed Ken is sufficient to downregulate a subset of JAK/STAT pathway target genes in vivo. Finally, we show that endogenous Ken specifically represses JAK/STAT-dependent expression of ventral veins lacking (vvl) in the posterior spiracles. Ken therefore represents a novel regulator of JAK/STAT signaling whose dynamic spatial and temporal expression is capable of selectively modulating the transcriptional repertoire elicited by activated STAT92E in vivo.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Tirosina Quinasas/metabolismo , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Drosophila/embriología , Drosophila/metabolismo , Proteínas de Drosophila/genética , Genes Reporteros , Quinasas Janus , Factores del Dominio POU/metabolismo , Filogenia , Homología de Secuencia de Aminoácido , Transducción de Señal
18.
J Leukoc Biol ; 106(5): 1063-1068, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31313387

RESUMEN

Methotrexate (MTX) is recognized as the anchor drug in the algorithm treating chronic arthritis (RA, psoriatic arthritis), as well as a steroid sparing agent in other inflammatory conditions (polymyalgia rheumatica, vasculitis, scleroderma). Its main mechanism of action has been related to the increase in extracellular adenosine, which leads to the effects of A2A receptor in M1 macrophages that dampens TNFα and IL12 production and increases IL1Ra and TNFRp75. By acting on A2B receptor on M2 macrophages it enhances IL10 synthesis and inhibits NF-kB signaling. MTX has also been shown to exert JAK inhibition of JAK2 and JAK1 when tested in Drosophila melanogaster as a model of kinase activity and in human cell lines (nodular sclerosis Hodgkin's lymphoma and acute myeloid leukemia cell lines). These effects may explain why MTX leads to clinical effects similar to anti-TNFα biologics in monotherapy, but is less effective when compared to anti-IL6R in monotherapy, which acting upstream exerts major effects downstream on the JAK1-STAT3 pathway. The MTX effects on JAK1/JAK2 inhibition also allows to understand why the combination of MTX with Leflunomide, or JAK1/JAK3 inhibitor leads to better clinical outcomes than monotherapy, while the combination with JAK1/JAK2 or JAK1 specific inhibitors does not seem to exert additive clinical benefit.


Asunto(s)
Artritis Psoriásica/tratamiento farmacológico , Inhibidores de las Cinasas Janus/uso terapéutico , Leflunamida/uso terapéutico , Metotrexato/uso terapéutico , Animales , Artritis Psoriásica/inmunología , Artritis Psoriásica/mortalidad , Quimioterapia Combinada , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/inmunología , Fiebre Reumática/inmunología , Fiebre Reumática/patología , Factor de Transcripción STAT3/inmunología
19.
Genetics ; 172(3): 1683-97, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16387886

RESUMEN

The JAK/STAT pathway was first identified in mammals as a signaling mechanism central to hematopoiesis and has since been shown to exert a wide range of pleiotropic effects on multiple developmental processes. Its inappropriate activation is also implicated in the development of numerous human malignancies, especially those derived from hematopoietic lineages. The JAK/STAT signaling cascade has been conserved through evolution and although the pathway identified in Drosophila has been closely examined, the full complement of genes required to correctly transduce signaling in vivo remains to be identified. We have used a dosage-sensitive dominant eye overgrowth phenotype caused by ectopic activation of the JAK/STAT pathway to screen 2267 independent, newly generated mutagenic P-element insertions. After multiple rounds of retesting, 23 interacting loci that represent genes not previously known to interact with JAK/STAT signaling have been identified. Analysis of these genes has identified three signal transduction pathways, seven potential components of the pathway itself, and six putative downstream pathway target genes. The use of forward genetics to identify loci and reverse genetic approaches to characterize them has allowed us to assemble a collection of genes whose products represent novel components and regulators of this important signal transduction cascade.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Janus Quinasa 1/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/genética , Animales , Proteínas de Ciclo Celular/genética , Drosophila/crecimiento & desarrollo , Drosophila/fisiología , Proteínas de Drosophila/fisiología , Ojo/crecimiento & desarrollo , Femenino , Regulación de la Expresión Génica/fisiología , Marcadores Genéticos , Hibridación in Situ , Janus Quinasa 1/fisiología , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Masculino , Mutagénesis Insercional , Interferencia de ARN , Factores de Transcripción STAT/fisiología
20.
Cell Signal ; 18(6): 819-29, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16129580

RESUMEN

In the canonical model of JAK/STAT signalling STAT transcription factors are activated by JAK mediated tyrosine phosphorylation following pathway stimulation by external cytokines. Activated STAT molecules then homo- or heterodimerise before translocating to the nucleus where they bind to DNA sequences within the promoters of pathway target genes. DNA-bound STAT dimers then activate transcription of their targets via interaction with components of the basal transcription machinery. Here we describe a missense mutation in the SH2 domain of the single Drosophila STAT92E homologue which results in an amino-acid substitution conserved in both the canonical SH2 domain and STAT-like molecules previously identified in C. elegans and the mosquito Anopheles gambiae. This mutation leads to nuclear accumulation and constitutive DNA binding of Drosophila STAT92E even in the absence of JAK stimulation. Strikingly, this mutant shows only limited transcriptional activity in tissue culture based assays and functions as a dominant-negative at both the phenotypic and molecular levels in vivo. These features represent aspects of both dominant gain-of-function and dominant-negative activities and imply that the functions of DNA binding can be functionally separated from the role of STAT92E as a transcriptional activator. It is thus possible that an alternative post-translational modification, in addition to tyrosine phosphorylation, may be required to allow STAT to act as a transcriptional activator and suggests the existence of an alternative mechanism by which STAT transcriptional activity may be regulated in vivo.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Mutación Missense , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Activación Transcripcional , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células Cultivadas , Análisis Mutacional de ADN , Drosophila , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Alineación de Secuencia , Dominios Homologos src/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda