Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35005772

RESUMEN

Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, our understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, we have monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). We show that the scrib mutation impacts the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch-dedicated transcription factor. The Notch-dependent neoplasms require, however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1 and basic leucine zipper factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally, our work highlights some Notch/scrib specificities, in particular the role of the PAR domain-containing basic leucine zipper transcription factor and Notch direct target Pdp1 for neoplastic growth.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Receptores Notch/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Carcinogénesis , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Larva/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Neoplasias/metabolismo , Neoplasias/patología , Interferencia de ARN , Transducción de Señal , Alas de Animales/metabolismo
2.
Curr Biol ; 30(10): 1809-1820.e4, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32275875

RESUMEN

Disease progression in many tumor types involves the interaction of genetically abnormal cancer cells with normal stromal cells. Neoplastic transformation in a Drosophila genetic model of epidermal growth factor receptor (EGFR)-driven tumorigenesis similarly relies on the interaction between epithelial and mesenchymal cells, providing a simple system to investigate mechanisms used for the cross-talk. Using the Drosophila model, we show that the transformed epithelium hijacks the mesenchymal cells through Notch signaling, which prevents their differentiation and promotes proliferation. A key downstream target in the mesenchyme is Zfh1/ZEB. When Notch or zfh1 are depleted in the mesenchymal cells, tumor growth is compromised. The ligand Delta is highly upregulated in the epithelial cells where it is found on long cellular processes. By using a live transcription assay in cultured cells and by depleting actin-rich processes in the tumor epithelium, we provide evidence that signaling can be mediated by cytonemes from Delta-expressing cells. We, thus, propose that high Notch activity in the unmodified mesenchymal cells is driven by ligands produced by the cancerous epithelial. This long-range Notch signaling integrates the two tissues to promote tumorigenesis, by co-opting a normal regulatory mechanism that prevents the mesenchymal cells from differentiating.


Asunto(s)
Carcinogénesis/metabolismo , Transformación Celular Neoplásica/metabolismo , Receptores Notch/metabolismo , Animales , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mioblastos/metabolismo , Receptores Notch/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal
3.
J Am Coll Cardiol ; 75(6): 565-574, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32057369

RESUMEN

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 inhibitor therapy is a treatment option for patients with familial hypercholesterolemia (FH) who are unable to reach low-density lipoprotein cholesterol (LDL-C) goals. OBJECTIVES: The aim of this study was to provide long-term safety and efficacy data for evolocumab in patients with homozygous FH (HoFH) and severe heterozygous FH (HeFH). METHODS: In this open-label, single-arm study, patients with HoFH or severe HeFH ≥12 years of age and on stable lipid-lowering therapy began subcutaneous evolocumab 420 mg monthly or 420 mg every 2 weeks if on lipoprotein apheresis. After 12 weeks, those not on apheresis could be up-titrated to 420 mg every 2 weeks. The primary endpoint was the incidence of treatment-emergent adverse events; secondary endpoints were changes in LDL-C and other lipids. RESULTS: In total, 300 patients (106 with HoFH, including 14 <18 years of age at enrollment) received evolocumab for a median of 4.1 years. Adverse events occurred in 89.3% of patients, the most common of which were nasopharyngitis, influenza, upper respiratory tract infection, and headache. Mean change in LDL-C from baseline to week 12 was -21.2% (-59.8 mg/dl) in patients with HoFH and -54.9% (-104.4 mg/dl) in those with severe HeFH and was sustained over time. Of 48 patients with HoFH who were up-titrated, mean change in LDL-C improved from -19.6% at week 12 to -29.7% after 12 weeks of 420 mg every 2 weeks. The adjudicated cardiovascular event rate was 2.7% per year. Of 61 patients receiving apheresis at enrollment, 16 discontinued apheresis. CONCLUSIONS: Evolocumab was well tolerated and effectively reduced plasma LDL-C levels in patients with HoFH and severe HeFH over a median of 4.1 years.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticolesterolemiantes/administración & dosificación , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticolesterolemiantes/efectos adversos , LDL-Colesterol/sangre , Femenino , Humanos , Hiperlipoproteinemia Tipo II/sangre , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Sci Rep ; 9(1): 18628, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819141

RESUMEN

In all metazoans, a small number of evolutionarily conserved signaling pathways are reiteratively used during development to orchestrate critical patterning and morphogenetic processes. Among these, Notch (N) signaling is essential for most aspects of tissue patterning where it mediates the communication between adjacent cells to control cell fate specification. In Drosophila, Notch signaling is required for several features of eye development, including the R3/R4 cell fate choice and R7 specification. Here we show that hypomorphic alleles of Notch, belonging to the Nfacet class, reveal a novel phenotype: while photoreceptor specification in the mutant ommatidia is largely normal, defects are observed in ommatidial rotation (OR), a planar cell polarity (PCP)-mediated cell motility process. We demonstrate that during OR Notch signaling is specifically required in the R4 photoreceptor to upregulate the transcription of argos (aos), an inhibitory ligand to the epidermal growth factor receptor (EGFR), to fine-tune the activity of EGFR signaling. Consistently, the loss-of-function defects of Nfacet alleles and EGFR-signaling pathway mutants are largely indistinguishable. A Notch-regulated aos enhancer confers R4 specific expression arguing that aos is directly regulated by Notch signaling in this context via Su(H)-Mam-dependent transcription.


Asunto(s)
Proteínas de Drosophila/genética , Receptores ErbB/genética , Proteínas del Ojo/genética , Proteínas del Tejido Nervioso/genética , Receptores de Péptidos de Invertebrados/genética , Receptores Notch/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Polaridad Celular/genética , Drosophila melanogaster/genética , Ojo/metabolismo , Receptores Frizzled/genética , Morfogénesis/genética , Células Fotorreceptoras de Invertebrados/metabolismo
6.
Elife ; 82019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30694174

RESUMEN

To progress towards differentiation, progeny of stem cells need to extinguish expression of stem-cell maintenance genes. Failures in such mechanisms can drive tumorigenesis. In Drosophila neural stem cell (NSC) lineages, excessive Notch signalling results in supernumerary NSCs causing hyperplasia. However, onset of hyperplasia is considerably delayed implying there are mechanisms that resist the mitogenic signal. Monitoring the live expression of a Notch target gene, E(spl)mγ, revealed that normal attenuation is still initiated in the presence of excess Notch activity so that re-emergence of NSC properties occurs only in older progeny. Screening for factors responsible, we found that depletion of Mi-2/NuRD ATP remodeling complex dramatically enhanced Notch-induced hyperplasia. Under these conditions, E(spl)mγ was no longer extinguished in NSC progeny. We propose that Mi-2 is required for decommissioning stem-cell enhancers in their progeny, enabling the switch towards more differentiated fates and rendering them insensitive to mitogenic factors such as Notch.


Asunto(s)
Adenosina Trifosfatasas/genética , Autoantígenos/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Hiperplasia/genética , Células-Madre Neurales/metabolismo , Receptores Notch/genética , Proteínas Represoras/genética , Adenosina Trifosfatasas/metabolismo , Animales , Autoantígenos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Linaje de la Célula/genética , Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Hiperplasia/metabolismo , Hiperplasia/patología , Modelos Biológicos , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal
8.
Dis Model Mech ; 10(4): 399-407, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28237966

RESUMEN

Myeloproliferative neoplasms (MPNs) of the Philadelphia-negative class comprise polycythaemia vera, essential thrombocythaemia and primary myelofibrosis (PMF). They are associated with aberrant numbers of myeloid lineage cells in the blood, and in the case of overt PMF, with development of myelofibrosis in the bone marrow and failure to produce normal blood cells. These diseases are usually caused by gain-of-function mutations in the kinase JAK2. Here, we use Drosophila to investigate the consequences of activation of the JAK2 orthologue in haematopoiesis. We have identified maturing haemocytes in the lymph gland, the major haematopoietic organ in the fly, as the cell population susceptible to induce hypertrophy upon targeted overexpression of JAK. We show that JAK activates a feed-forward loop, including the cytokine-like ligand Upd3 and its receptor, Domeless, which are required to induce lymph gland hypertrophy. Moreover, we present evidence that p38 MAPK signalling plays a key role in this process by inducing expression of the ligand Upd3. Interestingly, we also show that forced activation of the p38 MAPK pathway in maturing haemocytes suffices to generate hypertrophic organs and the appearance of melanotic tumours. Our results illustrate a novel pro-tumourigenic crosstalk between the p38 MAPK pathway and JAK signalling in a Drosophila model of MPNs. Based on the shared molecular mechanisms underlying MPNs in flies and humans, the interplay between Drosophila JAK and p38 signalling pathways unravelled in this work might have translational relevance for human MPNs.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas , Trastornos Mieloproliferativos/patología , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Modelos Animales de Enfermedad , Hemocitos/metabolismo , Hipertrofia , Ganglios Linfáticos/metabolismo
9.
Nat Rev Mol Cell Biol ; 17(11): 722-735, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27507209

RESUMEN

The highly conserved Notch signalling pathway functions in many different developmental and homeostatic processes, which raises the question of how this pathway can achieve such diverse outcomes. With a direct route from the membrane to the nucleus, the Notch pathway has fewer opportunities for regulation than do many other signalling pathways, yet it generates exquisitely patterned structures, including sensory hair cells and branched arterial networks. More confusingly, its activity promotes tissue growth and cancers in some circumstances but cell death and tumour suppression in others. Many different regulatory mechanisms help to shape the activity of the Notch pathway, generating functional outputs that are appropriate for each context. These mechanisms include the receptor-ligand landscape, the tissue topology, the nuclear environment and the connectivity of the regulatory networks.


Asunto(s)
Receptores Notch/fisiología , Transducción de Señal , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Humanos , Morfogénesis , Neovascularización Fisiológica
10.
Open Biol ; 6(2): 150155, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26887408

RESUMEN

Glycolytic shift is a characteristic feature of rapidly proliferating cells, such as cells during development and during immune response or cancer cells, as well as of stem cells. It results in increased glycolysis uncoupled from mitochondrial respiration, also known as the Warburg effect. Notch signalling is active in contexts where cells undergo glycolytic shift. We decided to test whether metabolic genes are direct transcriptional targets of Notch signalling and whether upregulation of metabolic genes can help Notch to induce tissue growth under physiological conditions and in conditions of Notch-induced hyperplasia. We show that genes mediating cellular metabolic changes towards the Warburg effect are direct transcriptional targets of Notch signalling. They include genes encoding proteins involved in glucose uptake, glycolysis, lactate to pyruvate conversion and repression of the tricarboxylic acid cycle. The direct transcriptional upregulation of metabolic genes is PI3K/Akt independent and occurs not only in cells with overactivated Notch but also in cells with endogenous levels of Notch signalling and in vivo. Even a short pulse of Notch activity is able to elicit long-lasting metabolic changes resembling the Warburg effect. Loss of Notch signalling in Drosophila wing discs as well as in human microvascular cells leads to downregulation of glycolytic genes. Notch-driven tissue overgrowth can be rescued by downregulation of genes for glucose metabolism. Notch activity is able to support growth of wing during nutrient-deprivation conditions, independent of the growth of the rest of the body. Notch is active in situations that involve metabolic reprogramming, and the direct regulation of metabolic genes may be a common mechanism that helps Notch to exert its effects in target tissues.


Asunto(s)
Ciclo del Ácido Cítrico/genética , Regulación de la Expresión Génica , Glucólisis/genética , Receptores Notch/metabolismo , Animales , Sitios de Unión , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Metabolismo Energético/genética , Expresión Génica , Genes Reporteros , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas , Unión Proteica , Receptores Notch/genética , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Activación Transcripcional
11.
Development ; 143(2): 219-31, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26657768

RESUMEN

Notch signalling is involved in a multitude of developmental decisions and its aberrant activation is linked to many diseases, including cancers. One example is the neural stem cell tumours that arise from constitutive Notch activity in Drosophila neuroblasts. To investigate how hyperactivation of Notch in larval neuroblasts leads to tumours, we combined results from profiling the upregulated mRNAs and mapping the regions bound by the core Notch pathway transcription factor Su(H). This identified 246 putative direct Notch targets. These genes were highly enriched for transcription factors and overlapped significantly with a previously identified regulatory programme dependent on the proneural transcription factor Asense. Included were genes associated with the neuroblast maintenance and self-renewal programme that we validated as Notch regulated in vivo. Another group were the so-called temporal transcription factors, which have been implicated in neuroblast maturation. Normally expressed in specific time windows, several temporal transcription factors were ectopically expressed in the stem cell tumours, suggesting that Notch had reprogrammed their normal temporal regulation. Indeed, the Notch-induced hyperplasia was reduced by mutations affecting two of the temporal factors, which, conversely, were sufficient to induce mild hyperplasia on their own. Altogether, the results suggest that Notch induces neuroblast tumours by directly promoting the expression of genes that contribute to stem cell identity and by reprogramming the expression of factors that could regulate maturity.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Receptores Notch/metabolismo , Animales , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster , Receptores Notch/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Int J Hematol Oncol Stem Cell Res ; 9(3): 112-21, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26261695

RESUMEN

BACKGROUND: Allogeneic Hematopoietic Stem Cell Transplantation (HSCT) is a curative treatment option for many patients with Acute Myeloid Leukemia (AML); however, it can lead to complications of Graft-Versus-Host-Disease (GVHD) which can affect the quality of life and overall survival. The aim of this study was to assess the effects of both acute and chronic GVHD on survival rate in patients with AML who received HSCT. SUBJECTS AND METHODS: In a longitudinal study, 587 patients with AML who underwent bone marrow transplantation in Tehran-Iran between1991 and 2011 were recruited. All patient records were analyzed for the occurrence of adverse events including acute and chronic GVHD and leukemia relapse. Data were analyzed using Log-rank, Kaplan-Meier, Univariate and Multivariate Cox Regression models. RESULTS: The five-year overall survival (OS) was found to be 71.9% (95% CI: 67.40-76.41). Also there was a significant relationship between cGVHD and OS (P=0.001, HR = 0.476, 95%). Hazard of death in these patients was less than those who did not experience an occurrence of cGVHD and aGVHD (HR= 0.629, P= 0.078). A significant relationship between cGVHD and relapse was observed (P< 0.001) indicating that patients who developed cGVHD experienced a better survival rate. A significant relationship was also found between overall survival and aGVHD grade (P< 0.001). Hazard of death (HD) for cGVHD and relapse variables were estimated to be 0.554 and 3.869. DISCUSSION: This study is one of the largest studies (regarding the number of participants) done to date in the Middle East with quite a long duration (20 years). cGVHD appears to have a positive influence on survival rate in patients with AML who received HSCT. It is recommended that further studies investigate the underlying reason or mechanisms behind this.

13.
J Cell Biol ; 210(2): 303-18, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26169355

RESUMEN

Notch signaling is a major regulator of cell fate, proliferation, and differentiation. Like other signaling pathways, its activity is strongly influenced by intracellular trafficking. Besides contributing to signal activation and down-regulation, differential fluxes between trafficking routes can cause aberrant Notch pathway activation. Investigating the function of the retromer-associated DNAJ protein Rme-8 in vivo, we demonstrate a critical role in regulating Notch receptor recycling. In the absence of Rme-8, Notch accumulated in enlarged tubulated Rab4-positive endosomes, and as a consequence, signaling was compromised. Strikingly, when the retromer component Vps26 was depleted at the same time, Notch no longer accumulated and instead was ectopically activated. Likewise, depletion of ESCRT-0 components Hrs or Stam in combination with Rme-8 also led to high levels of ectopic Notch activity. Together, these results highlight the importance of Rme-8 in coordinating normal endocytic recycling route and reveal that its absence predisposes toward conditions in which pathological Notch signaling can occur.


Asunto(s)
Proteínas de Drosophila/metabolismo , Receptores Notch/metabolismo , Animales , Desintegrinas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Técnicas de Silenciamiento del Gen , Metaloendopeptidasas/metabolismo , Transporte de Proteínas , Transducción de Señal , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteína Wnt1/metabolismo
14.
J Cell Sci ; 127(Pt 21): 4634-44, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25217625

RESUMEN

There is growing evidence that activation of the Notch pathway can result in consequences on cell morphogenesis and behaviour, both during embryonic development and cancer progression. In general, Notch is proposed to coordinate these processes by regulating expression of key transcription factors. However, many Notch-regulated genes identified in genome-wide studies are involved in fundamental aspects of cell behaviour, suggesting a more direct influence on cellular properties. By testing the functions of 25 such genes we confirmed that 12 are required in developing adult muscles, consistent with roles downstream of Notch. Focusing on three, Reck, rhea/talin and trio, we verify their expression in adult muscle progenitors and identify Notch-regulated enhancers in each. Full activity of these enhancers requires functional binding sites for Su(H), the DNA-binding transcription factor in the Notch pathway, validating their direct regulation. Thus, besides its well-known roles in regulating the expression of cell-fate-determining transcription factors, Notch signalling also has the potential to directly affect cell morphology and behaviour by modulating expression of genes such as Reck, rhea/talin and trio. This sheds new light on the functional outputs of Notch activation in morphogenetic processes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Células Madre/metabolismo , Talina/metabolismo , Animales , Drosophila , Proteínas de Drosophila/genética , Factores de Intercambio de Guanina Nucleótido/genética , Hibridación in Situ , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculos/citología , Músculos/metabolismo , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Talina/genética
15.
Apoptosis ; 19(10): 1430-43, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24858703

RESUMEN

A balance between cell proliferation and apoptosis is important for normal development and tissue homeostasis. Under stress conditions, the conserved tumor suppressor and transcription factor Dp53 induces apoptosis to contribute to the maintenance of homeostasis. However, in some cases Dp53-induced apoptosis results in the proliferation of surrounding non-apoptotic cells. To gain insight into the Dp53 function in the control of apoptosis and proliferation, we studied the interaction between the Drosophila Dp53 and Notch genes. We present evidence that simultaneous reduction of Dp53 and Notch function synergistically increases the wing phenotype of Notch heterozygous mutant flies. Further, we found that a Notch cis-regulatory element is responsive to loss and gain of Dp53 function and that over-expression of Dp53 up-regulates Notch mRNA and protein expression. These findings suggest not only that Dp53 and Notch act together to control wing development but also indicate that Dp53 transcriptionally regulates Notch expression. Moreover, using Notch  gain and loss of function mutations we examined the relevance of Dp53 and Notch interactions in the process of Dp53-apoptosis induced proliferation. Results show that proliferation induced by Dp53 over-expression is dependent on Notch, thus identifying Notch as a new player in Dp53-induced proliferation. Interestingly, we found that Dp53-induced Notch activation and proliferation occurs even under conditions where apoptosis was inhibited. Our findings highlight the conservation between flies and vertebrates of the Dp53 and Notch cross-talk and suggest that Dp53 has a dual role regulating cell death and proliferation gene networks to control the homeostatic balance between apoptosis and proliferation.


Asunto(s)
Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Receptores Notch/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Masculino , Receptores Notch/genética , Proteína p53 Supresora de Tumor/genética
16.
Stat Biopharm Res ; 6(1): 67-77, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24683441

RESUMEN

The objectives of the phase 2 stage in a drug development program are to evaluate the safety and tolerability of different doses, select a promising dose range, and look for early signs of activity. At the end of phase 2, a decision to initiate phase 3 studies is made that involves the commitment of considerable resources. This multifactorial decision, generally made by balancing the current condition of a development organization's portfolio, the future cost of development, the competitive landscape, and the expected safety and efficacy benefits of a new therapy, needs to be a good one. In this article, we present a practical quantitative process that has been implemented for drugs entering phase 2 at Amgen Ltd. to ensure a consistent and explicit evidence-based approach is used to contribute to decisions for new drug candidates. Broadly following this process will also help statisticians increase their strategic influence in drug development programs. The process is illustrated using an example from the pancreatic cancer indication. Embedded within the process is a predominantly Bayesian approach to predicting the probability of efficacy success in a future (frequentist) phase 3 program.

17.
Methods ; 68(1): 173-82, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24704358

RESUMEN

Notch signaling involves a highly conserved pathway that mediates communication between neighboring cells. Activation of Notch by its ligands, results in the release of the Notch intracellular domain (NICD), which enters the nucleus and regulates transcription. This pathway has been implicated in many developmental decisions and diseases (including cancers) over the past decades. The simplicity of the Notch pathway in Drosophila melanogaster, in combination with the availability of powerful genetics, make this an attractive model for studying fundamental principles of Notch regulation and function. In this article we present some of the established and emerging tools that are available to monitor and manipulate the Notch pathway in Drosophila and discuss their strengths and weaknesses.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Receptores Notch/genética , Transducción de Señal , Animales , Proteínas de Drosophila/metabolismo , Ligandos , Biología Molecular/métodos , Receptores Notch/metabolismo
18.
Nat Genet ; 46(1): 33-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24316979

RESUMEN

A major challenge in cancer genetics is to determine which low-frequency somatic mutations are drivers of tumorigenesis. Here we interrogate the genomes of 7,651 diverse human cancers and find inactivating mutations in the homeodomain transcription factor gene CUX1 (cut-like homeobox 1) in ~1-5% of various tumors. Meta-analysis of CUX1 mutational status in 2,519 cases of myeloid malignancies reveals disruptive mutations associated with poor survival, highlighting the clinical significance of CUX1 loss. In parallel, we validate CUX1 as a bona fide tumor suppressor using mouse transposon-mediated insertional mutagenesis and Drosophila cancer models. We demonstrate that CUX1 deficiency activates phosphoinositide 3-kinase (PI3K) signaling through direct transcriptional downregulation of the PI3K inhibitor PIK3IP1 (phosphoinositide-3-kinase interacting protein 1), leading to increased tumor growth and susceptibility to PI3K-AKT inhibition. Thus, our complementary approaches identify CUX1 as a pan-driver of tumorigenesis and uncover a potential strategy for treating CUX1-mutant tumors.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Homeodominio/genética , Mutación , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Animales , Elementos Transponibles de ADN , Drosophila/genética , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Insercional , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Factores de Transcripción , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Cell Biol ; 34(4): 664-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24324007

RESUMEN

Wiring between signaling pathways differs according to context, as exemplified by interactions between Notch and epidermal growth factor receptor (EGFR) pathways, which are cooperative in some contexts but antagonistic in others. To investigate mechanisms that underlie different modes of cross talk, we have focused on argos, an EGFR pathway regulator in Drosophila melanogaster which is upregulated by Notch in adult muscle progenitors but is repressed in the wing. Results show that the alternate modes of cross talk depend on the engagement of enhancers with opposite regulatory logic, which are selected by context-determining factors. This is likely to be a general mechanism for enabling the wiring between these pathways to switch according to context.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas del Ojo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Alas de Animales/crecimiento & desarrollo , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Receptores ErbB/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Receptores Notch/genética , Alas de Animales/metabolismo
20.
Development ; 140(4): 926-37, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23325760

RESUMEN

The diverse functions of Notch signalling imply that it must elicit context-specific programmes of gene expression. With the aim of investigating how Notch drives cells to differentiate, we have used a genome-wide approach to identify direct Notch targets in Drosophila haemocytes (blood cells), where Notch promotes crystal cell differentiation. Many of the identified Notch-regulated enhancers contain Runx and GATA motifs, and we demonstrate that binding of the Runx protein Lozenge (Lz) is required for enhancers to be competent to respond to Notch. Functional studies of targets, such as klumpfuss (ERG/WT1 family) and pebbled/hindsight (RREB1 homologue), show that Notch acts both to prevent the cells adopting alternate cell fates and to promote morphological characteristics associated with crystal cell differentiation. Inappropriate activity of Klumpfuss perturbs the differentiation programme, resulting in melanotic tumours. Thus, by acting as a master regulator, Lz directs Notch to activate selectively a combination of target genes that correctly locks cells into the differentiation programme.


Asunto(s)
Diferenciación Celular/fisiología , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Hemocitos/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Drosophila , Técnica del Anticuerpo Fluorescente , Hemocitos/metabolismo , Luciferasas , Mutagénesis , Proteínas Nucleares/metabolismo , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA