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











Base de dados
Intervalo de ano de publicação
1.
Front Med (Lausanne) ; 11: 1285772, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38698784

RESUMO

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.

2.
Int J Mol Sci ; 24(16)2023 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-37629022

RESUMO

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.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Hiperplasia , Agressão , Repetição de Anquirina , Divisão Celular , Proteínas de Ligação a RNA
3.
Front Immunol ; 14: 1310117, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38283366

RESUMO

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.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Drosophila melanogaster/genética , Eferocitose , Macrófagos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
4.
J Gerontol A Biol Sci Med Sci ; 77(8): 1494-1502, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34137822

RESUMO

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.


Assuntos
Proteínas de Drosophila , Ácido Mevalônico , Animais , Linhagem Celular Tumoral , Drosophila , Proteínas de Drosophila/metabolismo , Fatores de Transcrição Forkhead , Imidazóis/farmacologia , Mamíferos , Ácido Mevalônico/metabolismo , Ácido Zoledrônico/farmacologia
5.
Biochem Soc Trans ; 48(2): 559-567, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32239204

RESUMO

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.


Assuntos
Antirreumáticos/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Inflamação/tratamento farmacológico , Metotrexato/farmacologia , Artrite Reumatoide/tratamento farmacológico , Linhagem Celular , Doença de Crohn/tratamento farmacológico , Ácido Fólico/metabolismo , Humanos , Janus Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Psoríase/tratamento farmacológico , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
7.
J Leukoc Biol ; 106(5): 1063-1068, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31313387

RESUMO

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.


Assuntos
Artrite Psoriásica/tratamento farmacológico , Inibidores de Janus Quinases/uso terapêutico , Leflunomida/uso terapêutico , Metotrexato/uso terapêutico , Animais , Artrite Psoriásica/imunologia , Artrite Psoriásica/mortalidade , Quimioterapia Combinada , Humanos , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/imunologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/imunologia , Febre Reumática/imunologia , Febre Reumática/patologia , Fator de Transcrição STAT3/imunologia
8.
J Biol Chem ; 293(25): 9570-9579, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29695508

RESUMO

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.


Assuntos
Carcinoma de Células Renais/patologia , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/patologia , MicroRNAs/genética , Proteínas de Ligação a RNA/metabolismo , Sítios de Ligação , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Estudos de Casos e Controles , Movimento Celular , Perfilação da Expressão Gênica , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Prognóstico , Proteínas de Ligação a RNA/genética , Células Tumorais Cultivadas
10.
PLoS One ; 10(7): e0130078, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26131691

RESUMO

BACKGROUND: The JAK/STAT pathway transduces signals from multiple cytokines and controls haematopoiesis, immunity and inflammation. In addition, pathological activation is seen in multiple malignancies including the myeloproliferative neoplasms (MPNs). Given this, drug development efforts have targeted the pathway with JAK inhibitors such as ruxolitinib. Although effective, high costs and side effects have limited its adoption. Thus, a need for effective low cost treatments remains. METHODS & FINDINGS: We used the low-complexity Drosophila melanogaster pathway to screen for small molecules that modulate JAK/STAT signalling. This screen identified methotrexate and the closely related aminopterin as potent suppressors of STAT activation. We show that methotrexate suppresses human JAK/STAT signalling without affecting other phosphorylation-dependent pathways. Furthermore, methotrexate significantly reduces STAT5 phosphorylation in cells expressing JAK2 V617F, a mutation associated with most human MPNs. Methotrexate acts independently of dihydrofolate reductase (DHFR) and is comparable to the JAK1/2 inhibitor ruxolitinib. However, cells treated with methotrexate still retain their ability to respond to physiological levels of the ligand erythropoietin. CONCLUSIONS: Aminopterin and methotrexate represent the first chemotherapy agents developed and act as competitive inhibitors of DHFR. Methotrexate is also widely used at low doses to treat inflammatory and immune-mediated conditions including rheumatoid arthritis. In this low-dose regime, folate supplements are given to mitigate side effects by bypassing the biochemical requirement for DHFR. Although independent of DHFR, the mechanism-of-action underlying the low-dose effects of methotrexate is unknown. Given that multiple pro-inflammatory cytokines signal through the pathway, we suggest that suppression of the JAK/STAT pathway is likely to be the principal anti-inflammatory and immunosuppressive mechanism-of-action of low-dose methotrexate. In addition, we suggest that patients with JAK/STAT-associated haematological malignancies may benefit from low-dose methotrexate treatments. While the JAK1/2 inhibitor ruxolitinib is effective, a £43,200 annual cost precludes widespread adoption. With an annual methotrexate cost of around £32, our findings represent an important development with significant future potential.


Assuntos
Drosophila melanogaster/metabolismo , Inibidores Enzimáticos/farmacologia , Janus Quinase 2/metabolismo , Metotrexato/farmacologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Drosophila melanogaster/genética , Janus Quinase 2/genética
11.
J Cell Sci ; 127(Pt 1): 101-10, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24163435

RESUMO

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.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Hematopoese/genética , Hemócitos/metabolismo , Janus Quinases/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição/genética , Proteínas rho de Ligação ao GTP/genética , Animais , Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hemócitos/patologia , Janus Quinases/metabolismo , Masculino , Pseudópodes/metabolismo , Pseudópodes/patologia , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
12.
Methods Mol Biol ; 967: 81-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296723

RESUMO

The JAK/STAT signaling pathway has essential roles in multiple developmental processes, including stem cell maintenance, immune responses, and cellular proliferation. As a result, it has been extensively studied in both vertebrate systems and lower complexity models, such as Drosophila. Given its connection with such a wide range of biological functions, it is no surprise that pathway misregulation is frequently associated with multiple human diseases including cancer. While the core components of the pathway, and a number of negative regulators, are well known and conserved in many organisms, more subtle levels of regulation and inter-pathway crosstalk are less well understood. With the emergence of RNA interference (RNAi) as a tool to knock down gene expression and so evaluate protein function, high-throughput screens have been developed to identify pathway regulators on a genome-wide scale. Here we discuss the approaches and methods employed thus far for identification of pathway regulators using RNAi in Drosophila. Furthermore, we discuss possible approaches for future screens and the significant potential for applying RNAi technology in vertebrate models.


Assuntos
Técnicas Genéticas , Janus Quinases/metabolismo , Interferência de RNA , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética , Animais , Linhagem Celular Tumoral , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Humanos , Transcrição Gênica/genética
13.
JAKSTAT ; 1(1): 34-43, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24058749

RESUMO

Both the core JAK-STAT pathway components and their in vivo roles have been widely conserved between vertebrates and invertebrate models such as Drosophila melanogaster. Misregulation of JAK-STAT pathway activity has also been identified as a key factor in the development of multiple human malignancies. Recently, whole genome RNA interference (RNAi) screens in cultured Drosophila cells have identified both positively and negatively acting JAK-STAT pathway regulators. Here, we describe the analysis of 73 human genes representing homologs of 56 Drosophila genes originally identified by genome-wide RNAi screening as regulators of JAK-STAT signaling. Using assays for human STAT1 and STAT3 protein levels and phosphorylation status, as well as assays measuring the expression of endogenous STAT1 and STAT3 transcriptional targets, we have tested siRNAs targeting these 73 human genes and have identified potential JAK-STAT pathway regulatory roles in 69 (95%) of these. The genes identified represent a wide range of human JAK-STAT pathway regulators and include genes not previously known to modulate this signaling cascade. These results underline the value of model system based approaches for the identification of pathway regulators and have led to the identification of loci whose misregulation may ultimately be implicated in JAK-STAT pathway-mediated human disease.

14.
EMBO Rep ; 11(3): 201-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20168330

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica , Neoplasias Hematológicas/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Transcrição Gênica , Animais , Proliferação de Células , Análise por Conglomerados , Drosophila melanogaster , Regulação da Expressão Gênica , Células HeLa , Neoplasias Hematológicas/patologia , Hemócitos/citologia , Humanos , Camundongos , RNA Interferente Pequeno/metabolismo
15.
Curr Opin Genet Dev ; 18(5): 455-60, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18840523

RESUMO

The JAK/STAT signal transduction pathway has traditionally been viewed as a cytokine-stimulated activator of gene expression consisting of a straightforward receptor/JAK kinase/STAT transcription factor cascade. Recent studies in Drosophila, have, however consistently identified a range of chromatin-remodelling factors as regulators of in vivo JAK/STAT signalling. Now, the detailed analysis of one of these, heterochromatin protein 1 (HP1), has provided an insight into an unexpected non-canonical in vivo role for STAT. In this model, unphosphorylated STATs associate with and maintain the stability of transcriptionally repressed heterochromatin--an effect countered by the recruitment of STAT to the canonical pathway. We examine the background of this new model and its implications for JAK/STAT pathway requirements in stem cell maintenance and cancer.


Assuntos
Núcleo Celular/metabolismo , Drosophila/metabolismo , Fatores de Transcrição STAT/metabolismo , Animais , Diferenciação Celular , Núcleo Celular/genética , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Modelos Genéticos , Fosforilação , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo
16.
Genetics ; 172(3): 1683-97, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16387886

RESUMO

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.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Janus Quinase 1/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética , Animais , Proteínas de Ciclo Celular/genética , Drosophila/crescimento & desenvolvimento , Drosophila/fisiologia , Proteínas de Drosophila/fisiologia , Olho/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica/fisiologia , Marcadores Genéticos , Hibridização In Situ , Janus Quinase 1/fisiologia , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Masculino , Mutagênese Insercional , Interferência de RNA , Fatores de Transcrição STAT/fisiologia
17.
Curr Biol ; 16(1): 80-8, 2006 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-16401426

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Drosophila/embriologia , Drosophila/metabolismo , Proteínas de Drosophila/genética , Genes Reporter , Janus Quinases , Fatores do Domínio POU/metabolismo , Filogenia , Homologia de Sequência de Aminoácidos , Transdução de Sinais
18.
Nature ; 436(7052): 871-5, 2005 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16094372

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Genômica , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Interferência de RNA , Transdução de Sinais , Transativadores/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Epistasia Genética , Genoma , Hemócitos/citologia , Hemócitos/enzimologia , Hemócitos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fenótipo , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras , Fator de Transcrição STAT1 , Transativadores/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
19.
Oncogene ; 24(15): 2503-11, 2005 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-15735706

RESUMO

The JAK/STAT signalling pathway mediates both antiproliferative responses following interferon stimulation and cellular proliferation in response to cytokines such as interleukins and growth factors. Central to these responses are the seven vertebrate STAT molecules, misregulation of which is implicated in a variety of malignancies. We have investigated the proliferative role of the single Drosophila STAT92E, part of the evolutionarily conserved JAK/STAT cascade. During second instar larval wing disc development pathway activity is both necessary and sufficient to promote proliferation of this epithelial cell type. However by later stages, endogenous STAT92E is stimulated by a noncannonical mechanism to exert pronounced antiproliferative effects. Ectopic canonical activation is sufficient to further decrease proliferation and leads to the premature arrest of cells in the G2 phase of the cell cycle. The single STAT92E present in Drosophila therefore mediates both proproliferative functions analogous to vertebrate interleukin-stimulated STAT3 and antiproliferative functions analogous to interferon-stimulated STAT1. Pro- and antiproliferative roles therefore represent ancestral activities conserved through evolution and subsequently assigned to distinct molecules.


Assuntos
Ciclo Celular/fisiologia , Proliferação de Células , Proteínas de Ligação a DNA/farmacologia , Proteínas de Drosophila/farmacologia , Drosophila/crescimento & desenvolvimento , Drosophila/fisiologia , Proteínas Tirosina Quinases/farmacologia , Transativadores/farmacologia , Fatores de Transcrição/farmacologia , Animais , Janus Quinases , Larva/genética , Larva/crescimento & desenvolvimento , Fatores de Transcrição STAT , Transdução de Sinais
20.
Mech Dev ; 117(1-2): 343-6, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12204282

RESUMO

The suppressor of cytokine signalling (SOCS) gene family was originally identified as an immediate early response to cytokine signalling and function as negative regulators of the Janus kinase (JAK)/signal tranducers and activators of transcription (STAT) signal transduction pathway [Krebs and Hilton, J. Cell Sci. 113 (2000) 2813; Starr and Hilton, Int. J. Biochem. Cell Biol. 30 (1998) 1081]. Although key components of the Drosophila JAK/STAT pathway have been identified [Brown et al., Curr. Biol. 11 (2001) 1700, reviewed in Zeidler et al., Oncogene 19 (2000) 2598], regulators of the pathway, and SOCS genes in particular, have not yet been characterised. Here we report the cloning of Drosophila SOCS36E and show its expression pattern during embryonic and imaginal disc development. SOCS36E is expressed in an essentially identical pattern to the Drosophila JAK/STAT pathway ligand unpaired (Upd). It is not expressed in upd mutant embryos and is upregulated in response to ectopic activation of the pathway during both embryonic and imaginal development.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Proteínas Repressoras/genética , Fatores de Transcrição , Animais , Clonagem Molecular , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Glicoproteínas/genética , Mutação , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA