Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Cell ; 36(3): 288-301.e14, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31526759

RESUMEN

Current statistical models for assessing hotspot significance do not properly account for variation in site-specific mutability, thereby yielding many false-positives. We thus (i) detail a Log-normal-Poisson (LNP) background model that accounts for this variability in a manner consistent with models of mutagenesis; (ii) use it to show that passenger hotspots arise from all common mutational processes; and (iii) apply it to a ∼10,000-patient cohort to nominate driver hotspots with far fewer false-positives compared with conventional methods. Overall, we show that many cancer hotspot mutations recurring at the same genomic site across multiple tumors are actually passenger events, recurring at inherently mutable genomic sites under no positive selection.


Asunto(s)
Carcinogénesis/genética , Genómica/métodos , Modelos Genéticos , Mutagénesis , Neoplasias/genética , Análisis Mutacional de ADN , Conjuntos de Datos como Asunto , Genes Supresores de Tumor , Humanos , Distribución de Poisson , Curva ROC , Selección Genética , Secuenciación del Exoma
3.
J Cell Biol ; 217(9): 3183-3201, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-29934311

RESUMEN

ARHGAP35 encoding p190A RhoGAP is a cancer-associated gene with a mutation spectrum suggestive of a tumor-suppressor function. In this study, we demonstrate that loss of heterozygosity for ARHGAP35 occurs in human tumors. We sought to identify tumor-suppressor capacities for p190A RhoGAP (p190A) and its paralog p190B in epithelial cells. We reveal an essential role for p190A and p190B to promote contact inhibition of cell proliferation (CIP), a function that relies on RhoGAP activity. Unbiased mRNA sequencing analyses establish that p190A and p190B modulate expression of genes associated with the Hippo pathway. Accordingly, we determine that p190A and p190B induce CIP by repressing YAP-TEAD-regulated gene transcription through activation of LATS kinases and inhibition of the Rho-ROCK pathway. Finally, we demonstrate that loss of a single p190 paralog is sufficient to elicit nuclear translocation of YAP and perturb CIP in epithelial cells cultured in Matrigel. Collectively, our data reveal a novel mechanism consistent with a tumor-suppressor function for ARHGAP35.


Asunto(s)
Proliferación Celular/fisiología , Inhibición de Contacto/fisiología , Células Epiteliales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neoplasias/patología , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Perros , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica/genética , Factores de Intercambio de Guanina Nucleótido/genética , Vía de Señalización Hippo , Humanos , Células de Riñón Canino Madin Darby , Neoplasias/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Señalizadoras YAP , Quinasas Asociadas a rho/metabolismo
4.
J Exp Med ; 214(10): 2875-2887, 2017 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-28878000

RESUMEN

Recent advances in single-cell, transcriptomic profiling have provided unprecedented access to investigate cell heterogeneity during tissue and organ development. In this study, we used massively parallel, single-cell RNA sequencing to define cell heterogeneity within the zebrafish kidney marrow, constructing a comprehensive molecular atlas of definitive hematopoiesis and functionally distinct renal cells found in adult zebrafish. Because our method analyzed blood and kidney cells in an unbiased manner, our approach was useful in characterizing immune-cell deficiencies within DNA-protein kinase catalytic subunit (prkdc), interleukin-2 receptor γ a (il2rga), and double-homozygous-mutant fish, identifying blood cell losses in T, B, and natural killer cells within specific genetic mutants. Our analysis also uncovered novel cell types, including two classes of natural killer immune cells, classically defined and erythroid-primed hematopoietic stem and progenitor cells, mucin-secreting kidney cells, and kidney stem/progenitor cells. In total, our work provides the first, comprehensive, single-cell, transcriptomic analysis of kidney and marrow cells in the adult zebrafish.


Asunto(s)
Hematopoyesis Extramedular/genética , Riñón/citología , ARN/genética , Pez Cebra/anatomía & histología , Animales , Animales Modificados Genéticamente , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Perfilación de la Expresión Génica , Hematopoyesis Extramedular/fisiología , Células Madre Hematopoyéticas , Riñón/metabolismo , Análisis de Secuencia de ARN , Pez Cebra/genética , Pez Cebra/metabolismo
5.
Nat Biotechnol ; 35(10): 951-959, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28892075

RESUMEN

Microsatellites (MSs) are tracts of variable-length repeats of short DNA motifs that exhibit high rates of mutation in the form of insertions or deletions (indels) of the repeated motif. Despite their prevalence, the contribution of somatic MS indels to cancer has been largely unexplored, owing to difficulties in detecting them in short-read sequencing data. Here we present two tools: MSMuTect, for accurate detection of somatic MS indels, and MSMutSig, for identification of genes containing MS indels at a higher frequency than expected by chance. Applying MSMuTect to whole-exome data from 6,747 human tumors representing 20 tumor types, we identified >1,000 previously undescribed MS indels in cancer genes. Additionally, we demonstrate that the number and pattern of MS indels can accurately distinguish microsatellite-stable tumors from tumors with microsatellite instability, thus potentially improving classification of clinically relevant subgroups. Finally, we identified seven MS indel driver hotspots: four in known cancer genes (ACVR2A, RNF43, JAK1, and MSH3) and three in genes not previously implicated as cancer drivers (ESRP1, PRDM2, and DOCK3).


Asunto(s)
Mutación INDEL/genética , Repeticiones de Microsatélite/genética , Neoplasias/genética , Exoma/genética , Genes Relacionados con las Neoplasias , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inestabilidad de Microsatélites , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Nature ; 547(7661): 55-60, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28658208

RESUMEN

Genomic analysis of tumours has led to the identification of hundreds of cancer genes on the basis of the presence of mutations in protein-coding regions. By contrast, much less is known about cancer-causing mutations in non-coding regions. Here we perform deep sequencing in 360 primary breast cancers and develop computational methods to identify significantly mutated promoters. Clear signals are found in the promoters of three genes. FOXA1, a known driver of hormone-receptor positive breast cancer, harbours a mutational hotspot in its promoter leading to overexpression through increased E2F binding. RMRP and NEAT1, two non-coding RNA genes, carry mutations that affect protein binding to their promoters and alter expression levels. Our study shows that promoter regions harbour recurrent mutations in cancer with functional consequences and that the mutations occur at similar frequencies as in coding regions. Power analyses indicate that more such regions remain to be discovered through deep sequencing of adequately sized cohorts of patients.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica/genética , Mutación , Regiones Promotoras Genéticas/genética , Estudios de Cohortes , Factores de Transcripción E2F/metabolismo , Exoma/genética , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Unión Proteica/genética , ARN Largo no Codificante/genética , Receptores de Estrógenos/antagonistas & inhibidores
7.
J Biol Chem ; 292(7): 2679-2689, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28007963

RESUMEN

The negative regulator of Rho family GTPases, p190A RhoGAP, is one of six mammalian proteins harboring so-called FF motifs. To explore the function of these and other p190A segments, we identified interacting proteins by tandem mass spectrometry. Here we report that endogenous human p190A, but not its 50% identical p190B paralog, associates with all 13 eIF3 subunits and several other translational preinitiation factors. The interaction involves the first FF motif of p190A and the winged helix/PCI domain of eIF3A, is enhanced by serum stimulation and reduced by phosphatase treatment. The p190A/eIF3A interaction is unaffected by mutating phosphorylated p190A-Tyr308, but disrupted by a S296A mutation, targeting the only other known phosphorylated residue in the first FF domain. The p190A-eIF3 complex is distinct from eIF3 complexes containing S6K1 or mammalian target of rapamycin (mTOR), and appears to represent an incomplete preinitiation complex lacking several subunits. Based on these findings we propose that p190A may affect protein translation by controlling the assembly of functional preinitiation complexes. Whether such a role helps to explain why, unique among the large family of RhoGAPs, p190A exhibits a significantly increased mutation rate in cancer remains to be determined.


Asunto(s)
Factor 3 de Iniciación Eucariótica/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Biosíntesis de Proteínas , Proteínas Represoras/metabolismo , Animales , Cromatografía de Afinidad , Factor 3 de Iniciación Eucariótica/química , Factor 3 de Iniciación Eucariótica/genética , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Células HeLa , Humanos , Ratones , Mutación Missense , Células 3T3 NIH , Unión Proteica , Proteínas Represoras/química , Proteínas Represoras/genética , Fracciones Subcelulares/metabolismo
8.
Nat Med ; 21(11): 1290-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26501192

RESUMEN

Genome-wide association studies (GWASs) have linked genes to various pathological traits. However, the potential contribution of regulatory noncoding RNAs, such as microRNAs (miRNAs), to a genetic predisposition to pathological conditions has remained unclear. We leveraged GWAS meta-analysis data from >188,000 individuals to identify 69 miRNAs in physical proximity to single-nucleotide polymorphisms (SNPs) associated with abnormal levels of circulating lipids. Several of these miRNAs (miR-128-1, miR-148a, miR-130b, and miR-301b) control the expression of key proteins involved in cholesterol-lipoprotein trafficking, such as the low-density lipoprotein (LDL) receptor (LDLR) and the ATP-binding cassette A1 (ABCA1) cholesterol transporter. Consistent with human liver expression data and genetic links to abnormal blood lipid levels, overexpression and antisense targeting of miR-128-1 or miR-148a in high-fat diet-fed C57BL/6J and Apoe-null mice resulted in altered hepatic expression of proteins involved in lipid trafficking and metabolism, and in modulated levels of circulating lipoprotein-cholesterol and triglycerides. Taken together, these findings support the notion that altered expression of miRNAs may contribute to abnormal blood lipid levels, predisposing individuals to human cardiometabolic disorders.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , Dieta Alta en Grasa , Dislipidemias/genética , MicroARNs/genética , Receptores de LDL/metabolismo , Triglicéridos/metabolismo , Animales , Apolipoproteínas E/genética , Colesterol/metabolismo , Estudio de Asociación del Genoma Completo , Homeostasis/genética , Humanos , Lipoproteínas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple
9.
Rare Dis ; 2: e28341, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25054093

RESUMEN

Neurofibromatosis type 1 (NF1) is caused by loss of a negative regulator of Ras oncoproteins. Unknown genetic modifiers have been implicated in NF1's characteristic variability. Drosophila melanogaster dNf1 phenotypes include cognitive deficits and reduced growth, both of which resemble human symptoms. We recently reported results of a screen for dominant modifiers of dNf1 growth. Suppressors include the dAlk tyrosine kinase and its activating ligand, two other genes involved in Ras/ERK signal transduction, the synaptic scaffold Dap160 and the CCKLR-17D1 drosulfakinin receptor. Additional modifiers include several genes involved in cAMP/PKA signaling. Providing mechanistic insights, dAlk, jeb, and CCKLR-17D1 also suppress a dNf1 synaptic overgrowth defect, and increasing cAMP/PKA signaling in the neuroendocrine ring gland rescued the dNf1 growth deficiency. Finally, among the several suppressors identified in our screen, we specifically implicate ALK as a potential therapeutic target by showing that NF1-regulated ALK/RAS/ERK signaling is conserved in human cells.

10.
Am J Med Genet A ; 164A(3): 563-78, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24443315

RESUMEN

The neurofibromatoses (NF) are autosomal dominant genetic disorders that encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect more people worldwide than Duchenne muscular dystrophy and Huntington's disease combined. NF1 and NF2 are caused by mutations of known tumor suppressor genes (NF1 and NF2, respectively). For schwannomatosis, although mutations in SMARCB1 were identified in a subpopulation of schwannomatosis patients, additional causative gene mutations are still to be discovered. Individuals with NF1 may demonstrate manifestations in multiple organ systems, including tumors of the nervous system, learning disabilities, and physical disfigurement. NF2 ultimately can cause deafness, cranial nerve deficits, and additional severe morbidities caused by tumors of the nervous system. Unmanageable pain is a key finding in patients with schwannomatosis. Although today there is no marketed treatment for NF-related tumors, a significant number of clinical trials have become available. In addition, significant preclinical efforts have led to a more rational selection of potential drug candidates for NF trials. An important element in fueling this progress is the sharing of knowledge. For over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share novel findings, ideas, and build collaborations. The 2012 NF Conference held in New Orleans hosted over 350 NF researchers and clinicians. This article provides a synthesis of the highlights presented at the conference and as such, is a "state-of-the-field" for NF research in 2012.


Asunto(s)
Neurilemoma/etiología , Neurofibromatosis/etiología , Neurofibromatosis 1/etiología , Neurofibromatosis 2/etiología , Neoplasias Cutáneas/etiología , Humanos , Neurilemoma/genética , Neurilemoma/terapia , Neurofibromatosis/genética , Neurofibromatosis/terapia , Neurofibromatosis 1/genética , Neurofibromatosis 1/terapia , Neurofibromatosis 2/genética , Neurofibromatosis 2/terapia , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia
11.
PLoS Genet ; 9(11): e1003958, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24278035

RESUMEN

Neurofibromatosis type 1 (NF1), a genetic disease that affects 1 in 3,000, is caused by loss of a large evolutionary conserved protein that serves as a GTPase Activating Protein (GAP) for Ras. Among Drosophila melanogaster Nf1 (dNf1) null mutant phenotypes, learning/memory deficits and reduced overall growth resemble human NF1 symptoms. These and other dNf1 defects are relatively insensitive to manipulations that reduce Ras signaling strength but are suppressed by increasing signaling through the 3'-5' cyclic adenosine monophosphate (cAMP) dependent Protein Kinase A (PKA) pathway, or phenocopied by inhibiting this pathway. However, whether dNf1 affects cAMP/PKA signaling directly or indirectly remains controversial. To shed light on this issue we screened 486 1(st) and 2(nd) chromosome deficiencies that uncover >80% of annotated genes for dominant modifiers of the dNf1 pupal size defect, identifying responsible genes in crosses with mutant alleles or by tissue-specific RNA interference (RNAi) knockdown. Validating the screen, identified suppressors include the previously implicated dAlk tyrosine kinase, its activating ligand jelly belly (jeb), two other genes involved in Ras/ERK signal transduction and several involved in cAMP/PKA signaling. Novel modifiers that implicate synaptic defects in the dNf1 growth deficiency include the intersectin-related synaptic scaffold protein Dap160 and the cholecystokinin receptor-related CCKLR-17D1 drosulfakinin receptor. Providing mechanistic clues, we show that dAlk, jeb and CCKLR-17D1 are among mutants that also suppress a recently identified dNf1 neuromuscular junction (NMJ) overgrowth phenotype and that manipulations that increase cAMP/PKA signaling in adipokinetic hormone (AKH)-producing cells at the base of the neuroendocrine ring gland restore the dNf1 growth deficiency. Finally, supporting our previous contention that ALK might be a therapeutic target in NF1, we report that human ALK is expressed in cells that give rise to NF1 tumors and that NF1 regulated ALK/RAS/ERK signaling appears conserved in man.


Asunto(s)
Drosophila melanogaster/genética , Trastornos de la Memoria/genética , Neurofibromatosis 1/genética , Quinasa de Linfoma Anaplásico , Animales , AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Trastornos de la Memoria/patología , Mutación , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/fisiopatología , Unión Neuromuscular/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
12.
PLoS Genet ; 7(9): e1002281, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21949657

RESUMEN

Anaplastic Lymphoma Kinase (Alk) is a Receptor Tyrosine Kinase (RTK) activated in several cancers, but with largely unknown physiological functions. We report two unexpected roles for the Drosophila ortholog dAlk, in body size determination and associative learning. Remarkably, reducing neuronal dAlk activity increased body size and enhanced associative learning, suggesting that its activation is inhibitory in both processes. Consistently, dAlk activation reduced body size and caused learning deficits resembling phenotypes of null mutations in dNf1, the Ras GTPase Activating Protein-encoding conserved ortholog of the Neurofibromatosis type 1 (NF1) disease gene. We show that dAlk and dNf1 co-localize extensively and interact functionally in the nervous system. Importantly, genetic or pharmacological inhibition of dAlk rescued the reduced body size, adult learning deficits, and Extracellular-Regulated-Kinase (ERK) overactivation dNf1 mutant phenotypes. These results identify dAlk as an upstream activator of dNf1-regulated Ras signaling responsible for several dNf1 defects, and they implicate human Alk as a potential therapeutic target in NF1.


Asunto(s)
Aprendizaje por Asociación , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Tamaño Corporal/genética , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Terapia Molecular Dirigida , Mutación , Proteínas del Tejido Nervioso/genética , Neurofibromina 1/antagonistas & inhibidores , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Proteínas Activadoras de ras GTPasa/genética
13.
Am J Med Genet A ; 155A(2): 307-21, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21271647

RESUMEN

The neurofibromatoses (NF) encompass the rare diseases NF1, NF2, and schwannomatosis. The NFs affect 100,000 Americans; over 2 million persons worldwide; and are caused by mutation of tumor suppressor genes. Individuals with NF1 in particular may develop tumors anywhere in the nervous system; additional manifestations can include learning disabilities, bone dysplasia, cardiovascular defects, unmanageable pain, and physical disfigurement. Ultimately, the NFs can cause blindness, deafness, severe morbidity, and increased mortality and NF1 includes a risk of malignant cancer. Today there is no treatment for the NFs (other than symptomatic); however, research efforts to understand these genetic conditions have made tremendous strides in the past few years. Progress is being made on all fronts, from discovery studies-understanding the molecular signaling deficits that cause the manifestations of NF-to the growth of preclinical drug screening initiatives and the emergence of a number of clinical trials. An important element in fuelling this progress is the sharing of knowledge, and to this end, for over 20 years the Children's Tumor Foundation has convened an annual NF Conference, bringing together NF professionals to share ideas and build collaborations. The 2010 NF Conference held in Baltimore, MD June 5-8, 2010 hosted over 300 NF researchers and clinicians. This paper provides a synthesis of the highlights presented at the Conference and as such, is a "state-of-the-field" for NF research in 2010.


Asunto(s)
Genes Supresores de Tumor , Neurofibromatosis/diagnóstico , Neurofibromatosis/tratamiento farmacológico , Neurofibromatosis/patología , Transducción de Señal/fisiología , Animales , Modelos Animales de Enfermedad , Genes ras/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neurofibromatosis/genética
15.
J Neurosci ; 27(17): 4716-24, 2007 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-17460084

RESUMEN

In the germinative zone of the adult rodent brain, neural progenitors migrate into niches delimited by astrocyte processes and differentiate into neuronal precursors. In the present study, we report a modulating role for the scaffolding protein IQGAP1 in neural progenitor migration. We have identified IQGAP1 as a new marker of amplifying neural progenitor and neuronal precursor cells of the subventricular zone (SVZ) and the rostral migratory stream (RMS) in the adult mouse brain. To determine functions for IQGAP1 in neural progenitors, we compared the properties of neural progenitor cells from wild-type and Iqgap1-null mutant mice in vivo and in vitro. The in vivo studies reveal a delay in the transition of de novo neural progenitors into neuronal precursor cells in Iqgap1-null mice. In vitro, we demonstrated that IQGAP1 acts as a downstream effector in the vascular endothelial growth factor (VEGF)-dependent migratory response of neural progenitors that also impacts on their neuronal differentiation. The Rho-family GTPases cdc42/Rac1 and Lis1 are major partners of IQGAP1 in this migratory process. Finally, astrocytes of the neurogenic SVZ and RMS are shown to express VEGF. We propose that VEGF synthesized by astrocytes could be involved in the guidance of neural progenitors to neurogenic niches and that IQGAP1 is an effector of the VEGF-dependent migratory signal.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Movimiento Celular/fisiología , Neuronas/citología , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Ventrículos Cerebrales/citología , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuropéptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Proteínas Activadoras de ras GTPasa/genética
17.
Growth Factors ; 25(5): 355-61, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18236214

RESUMEN

Evolutionary conserved members of the Ras superfamily of small GTP-binding proteins function as binary molecular switches to control diverse biological processes. In the context of cellular signaling, these include functions in exocytic and endocytic trafficking, as well as roles in signal relay downstream of various cell surface receptors. We previously reviewed roles played by the large family of GTPase, activating proteins in these processes. In this companion review, we highlight recent findings relating to the regulation of another major class of Ras superfamily regulatory proteins, the guanine nucleotide exchange factors.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Transducción de Señal/fisiología , Animales , Enfermedades Genéticas Congénitas/etiología , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Proteína SOS1/fisiología
18.
Genes Dev ; 20(23): 3311-23, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17114577

RESUMEN

Neurofibromatosis type 1 (NF1) is among the most common genetic disorders of humans and is caused by loss of neurofibromin, a large and highly conserved protein whose only known function is to serve as a GTPase-Activating Protein (GAP) for Ras. However, most Drosophila NF1 mutant phenotypes, including an overall growth deficiency, are not readily modified by manipulating Ras signaling strength, but are rescued by increasing signaling through the cAMP-dependent protein kinase A pathway. This has led to suggestions that NF1 has distinct Ras- and cAMP-related functions. Here we report that the Drosophila NF1 growth defect reflects a non-cell-autonomous requirement for NF1 in larval neurons that express the R-Ras ortholog Ras2, that NF1 is a GAP for Ras1 and Ras2, and that a functional NF1-GAP catalytic domain is both necessary and sufficient for rescue. Moreover, a Drosophila p120RasGAP ortholog, when expressed in the appropriate cells, can substitute for NF1 in growth regulation. Our results show that loss of NF1 can give rise to non-cell-autonomous developmental defects, implicate aberrant Ras-mediated signaling in larval neurons as the primary cause of the NF1 growth deficiency, and argue against the notion that neurofibromin has separable Ras- and cAMP-related functions.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/genética , Neurofibromina 1/genética , Neuronas/fisiología , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Dosificación de Gen , Larva , Mutación , Neuronas/enzimología , Supresión Genética , Proteínas Activadoras de ras GTPasa/genética
19.
Methods Enzymol ; 407: 1-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16757309

RESUMEN

For geneticists and other researchers alike it is often useful to know how many related proteins might perform similar functions. With this in mind, a survey was performed to determine what proportion of human and Drosophila genes code for Ras superfamily members and their positive or negative regulators. Results indicate that just < 2% of genes in both genomes predict such proteins. A database was compiled to provide easy access to this information. This database also includes information on approximately 360 putative Ras superfamily effector proteins and may be a useful tool for those interested in GTPase biology.


Asunto(s)
Bases de Datos de Proteínas , Proteínas Activadoras de ras GTPasa/genética , Proteínas ras/genética , Animales , Humanos , Proteínas de Unión al GTP Monoméricas/genética
20.
Growth Factors ; 23(2): 143-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16019436

RESUMEN

Approximately 2% of genes predicted by the sequenced human genome encode small GTPases and their regulators, highlighting the biological significance of regulated GTPase activity. Among the key GTPase regulators are the GTPase activating proteins (GAPs), which function to down-modulate active GTPases. Of the numerous identified GAPs, several have been implicated in signal transduction downstream of growth factors. In particular, GAPs for the Ras and Rho GTPases, which mediate a variety of receptor-transduced signals, appear to play an essential role in growth factor dependent GTPase regulation. Experimental studies of several of the GAPs have begun to elucidate mechanisms by which GAP activity is influenced by growth factor signaling, including direct phosphorylation, sub-cellular redistribution and protein degradation. Here, some of these mechanisms of GAP regulation in the context of signaling responses to growth factors are reviewed.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Regulación de la Expresión Génica , Sustancias de Crecimiento/metabolismo , Transducción de Señal , Encéfalo/metabolismo , Regulación hacia Abajo , Proteínas Activadoras de GTPasa/metabolismo , Genoma Humano , Genómica/métodos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Neurofibromina 1/metabolismo , Proteínas Nucleares/metabolismo , Fosfolípidos/metabolismo , Fosforilación , Proteómica/métodos , Proteínas Represoras/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , ras-GRF1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...