Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(24): e2400732121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38838021

RESUMEN

Cytoplasmic mislocalization and aggregation of TDP-43 protein are hallmarks of amyotrophic lateral sclerosis (ALS) and are observed in the vast majority of both familial and sporadic cases. How these two interconnected processes are regulated on a molecular level, however, remains enigmatic. Genome-wide screens for modifiers of the ALS-associated genes TDP-43 and FUS have identified the phospholipase D (Pld) pathway as a key regulator of ALS-related phenotypes in the fruit fly Drosophila melanogaster [M. W. Kankel et al., Genetics 215, 747-766 (2020)]. Here, we report the results of our search for downstream targets of the enzymatic product of Pld, phosphatidic acid. We identify two conserved negative regulators of the cAMP/PKA signaling pathway, the phosphodiesterase dunce and the inhibitory subunit PKA-R2, as modifiers of pathogenic phenotypes resulting from overexpression of the Drosophila TDP-43 ortholog TBPH. We show that knockdown of either of these genes results in a mitigation of both TBPH aggregation and mislocalization in larval motor neuron cell bodies, as well as an amelioration of adult-onset motor defects and shortened lifespan induced by TBPH. We determine that PKA kinase activity is downstream of both TBPH and Pld and that overexpression of the PKA target CrebA can rescue TBPH mislocalization. These findings suggest a model whereby increasing cAMP/PKA signaling can ameliorate the molecular and functional effects of pathological TDP-43.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico , AMP Cíclico , Proteínas de Unión al ADN , Proteínas de Drosophila , Drosophila melanogaster , Transducción de Señal , Animales , AMP Cíclico/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Humanos , Neuronas Motoras/metabolismo
2.
EMBO J ; 36(21): 3232-3249, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29030483

RESUMEN

Notch signaling is an evolutionarily conserved signal transduction pathway that is essential for metazoan development. Upon ligand binding, the Notch intracellular domain (NOTCH ICD) translocates into the nucleus and forms a complex with the transcription factor RBPJ (also known as CBF1 or CSL) to activate expression of Notch target genes. In the absence of a Notch signal, RBPJ acts as a transcriptional repressor. Using a proteomic approach, we identified L3MBTL3 (also known as MBT1) as a novel RBPJ interactor. L3MBTL3 competes with NOTCH ICD for binding to RBPJ In the absence of NOTCH ICD, RBPJ recruits L3MBTL3 and the histone demethylase KDM1A (also known as LSD1) to the enhancers of Notch target genes, leading to H3K4me2 demethylation and to transcriptional repression. Importantly, in vivo analyses of the homologs of RBPJ and L3MBTL3 in Drosophila melanogaster and Caenorhabditis elegans demonstrate that the functional link between RBPJ and L3MBTL3 is evolutionarily conserved, thus identifying L3MBTL3 as a universal modulator of Notch signaling in metazoans.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Histona Demetilasas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Neuroglía/metabolismo , Receptores Notch/genética , Animales , Evolución Biológica , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Histona Demetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Neuroglía/citología , Unión Proteica , Dominios Proteicos , Receptores Notch/metabolismo , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
3.
Adv Exp Med Biol ; 1066: 125-140, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30030825

RESUMEN

The Notch pathway controls a very broad spectrum of cell fates in metazoans during development, influencing proliferation, differentiation and cell death. Given its central role in normal development and homeostasis, misregulation of Notch signals can lead to various disorders including cancer. How the Notch pathway mediates such pleiotropic and differential effects is of fundamental importance. It is becoming increasingly clear through a number of large-scale genetic and proteomic studies that Notch interacts with a staggeringly large number of other genes and pathways in a context-dependent, complex, and highly regulated network, which determines the ultimate biological outcome. How best to interpret and analyze the continuously increasing wealth of data on Notch interactors remains a challenge. Here we review the current state of genetic and proteomic data related to the Notch interactome.


Asunto(s)
Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Receptores Notch/genética
4.
EMBO J ; 31(13): 2895-907, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22580825

RESUMEN

Genetic analyses in Drosophila revealed a synergy between Notch and the pleiotropic transcription factor Mef2 (myocyte enhancer factor 2), which profoundly influences proliferation and metastasis. We show that these hyperproliferative and invasive Drosophila phenotypes are attributed to upregulation of eiger, a member of the tumour necrosis factor superfamily of ligands, and the consequent activation of Jun N-terminal kinase signalling, which in turn triggers the expression of the invasive marker MMP1. Expression studies in human breast tumour samples demonstrate correlation between Notch and Mef2 paralogues and support the notion that Notch-MEF2 synergy may be significant for modulating human mammary oncogenesis.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factores Reguladores Miogénicos/fisiología , Receptores Notch/fisiología , Animales , Neoplasias de la Mama/metabolismo , Proteínas de Drosophila/biosíntesis , Femenino , Perfilación de la Expresión Génica , Humanos , Factores de Transcripción MEF2 , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Proteínas de la Membrana/biosíntesis , Factores Reguladores Miogénicos/metabolismo , Metástasis de la Neoplasia , Regulación hacia Arriba
5.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33902813

RESUMEN

Oncogenes often promote cell death as well as proliferation. How oncogenes drive these diametrically opposed phenomena remains to be solved. A key question is whether cell death occurs as a response to aberrant proliferation signals or through a proliferation-independent mechanism. Here, we reveal that Src, the first identified oncogene, simultaneously drives cell proliferation and death in an obligatorily coupled manner through parallel MAPK pathways. The two MAPK pathways diverge from a lynchpin protein Slpr. A MAPK p38 drives proliferation whereas another MAPK JNK drives apoptosis independently of proliferation signals. Src-p38-induced proliferation is regulated by methionine-mediated Tor signaling. Reduction of dietary methionine uncouples the obligatory coupling of cell proliferation and death, suppressing tumorigenesis and tumor-induced lethality. Our findings provide an insight into how cells evolved to have a fail-safe mechanism that thwarts tumorigenesis by the oncogene Src. We also exemplify a diet-based approach to circumvent oncogenesis by exploiting the fail-safe mechanism.


Asunto(s)
Muerte Celular , Proliferación Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Metionina/deficiencia , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Larva/genética , Larva/crecimiento & desarrollo , Larva/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
6.
Wiley Interdiscip Rev Dev Biol ; 9(1): e358, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31502763

RESUMEN

The role of the Notch signaling pathway in neural development has been well established over many years. More recent studies, however, have demonstrated that Notch continues to be expressed and active throughout adulthood in many areas of the central nervous system. Notch signals have been implicated in adult neurogenesis, memory formation, and synaptic plasticity in the adult organism, as well as linked to acute brain trauma and chronic neurodegenerative conditions. NOTCH3 mutations are responsible for the most common form of hereditary stroke, the progressive disorder cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Notch has also been associated with several progressive neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and amyotrophic lateral sclerosis. Although numerous studies link Notch activity with CNS homeostasis and neurodegenerative diseases, the data thus far are primarily correlative, rather than functional. Nevertheless, the evidence for Notch pathway activity in specific neural cellular contexts is strong, and certainly intriguing, and points to the possibility that the pathway carries therapeutic promise. This article is categorized under: Nervous System Development > Flies Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: General Principles.


Asunto(s)
Sistema Nervioso Central/metabolismo , Homeostasis/fisiología , Enfermedades Neurodegenerativas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Mutación/fisiología
7.
Dev Biol ; 315(1): 203-16, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18234181

RESUMEN

Xenopus tadpoles can fully regenerate all major tissue types following tail amputation. TGF-beta signaling plays essential roles in growth, repair, specification, and differentiation of tissues throughout development and adulthood. We examined the localization of key components of the TGF-beta signaling pathway during regeneration and characterized the effects of loss of TGF-beta signaling on multiple regenerative events. Phosphorylated Smad2 (p-Smad2) is initially restricted to the p63+ basal layer of the regenerative epithelium shortly after amputation, and is later found in multiple tissue types in the regeneration bud. TGF-beta ligands are also upregulated throughout regeneration. Treatment of amputated tails with SB-431542, a specific and reversible inhibitor of TGF-beta signaling, blocks tail regeneration at multiple points. Inhibition of TGF-beta signaling immediately following tail amputation reversibly prevents formation of a wound epithelium over the future regeneration bud. Even brief inhibition immediately following amputation is sufficient, however, to irreversibly block the establishment of structures and cell types that characterize regenerating tissue and to prevent the proper activation of BMP and ERK signaling pathways. Inhibition of TGF-beta signaling after regeneration has already commenced blocks cell proliferation in the regeneration bud. These data reveal several spatially and temporally distinct roles for TGF-beta signaling during regeneration: (1) wound epithelium formation, (2) establishment of regeneration bud structures and signaling cascades, and (3) regulation of cell proliferation.


Asunto(s)
Regeneración/fisiología , Transducción de Señal , Cola (estructura animal)/fisiología , Factor de Crecimiento Transformador beta/fisiología , Xenopus/fisiología , Animales , Benzamidas/farmacología , Dioxoles/farmacología , Regulación de la Expresión Génica , Histocitoquímica , Hibridación in Situ , Cinética , Larva , Ligandos , Fosforilación , Proteína Smad2/genética , Proteína Smad2/fisiología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología
8.
Curr Top Dev Biol ; 116: 17-33, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26970611

RESUMEN

The essential and highly conserved Notch signaling pathway controls a wide range of cell fate decisions during development, including cellular proliferation. Notch mediates both pro- and anti-proliferative effects in development, stem cells, and cancer depending on cellular context. Furthermore, it can induce proliferation in both cell-autonomous and non-cell-autonomous manners. Interacting genes and crosstalking signaling pathways play essential roles in regulating the proliferative response to Notch signals. A large number of genes that participate in the Notch network to influence proliferation have been identified, including several that activate the JNK signaling pathway, which interacts with Notch to induce both hyperplastic and invasive cellular behaviors. It is clear that dissecting the genetic circuitry surrounding Notch is essential to understanding the proliferative response to Notch in both development and cancer.


Asunto(s)
Neoplasias/patología , Receptores Notch/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Drosophila/embriología , Drosophila/genética , Drosophila/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Neoplasias/metabolismo , Receptores Notch/genética , Transducción de Señal/fisiología
9.
Elife ; 4: e05996, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26222204

RESUMEN

Notch signaling controls a wide range of cell fate decisions during development and disease via synergistic interactions with other signaling pathways. Here, through a genome-wide genetic screen in Drosophila, we uncover a highly complex Notch-dependent genetic circuitry that profoundly affects proliferation and consequently hyperplasia. We report a novel synergistic relationship between Notch and either of the non-receptor tyrosine kinases Src42A and Src64B to promote hyperplasia and tissue disorganization, which results in cell cycle perturbation, JAK/STAT signal activation, and differential regulation of Notch targets. Significantly, the JNK pathway is responsible for the majority of the phenotypes and transcriptional changes downstream of Notch-Src synergy. We previously reported that Notch-Mef2 also activates JNK, indicating that there are commonalities within the Notch-dependent proliferation circuitry; however, the current data indicate that Notch-Src accesses JNK in a significantly different fashion than Notch-Mef2.


Asunto(s)
Proliferación Celular , Proteínas de Drosophila/metabolismo , Hiperplasia , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Animales , Drosophila
10.
Mech Dev ; 127(9-12): 485-95, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20807570

RESUMEN

A key role for phosphorylation of Smad2 by TGFß superfamily ligands in the axial patterning of early embryos is well established. The regulation and role of Smad2 signaling in post-neurula embryonic patterning, however, is less well understood. While a variety of TGFß superfamily ligands are implicated in various stages of anterior-posterior patterning, the ligand GDF11 has been shown to have a particular role in post-gastrula patterning in the mouse. Mouse GDF11 is specifically localized to the developing tail and is essential for normal posterior axial patterning. Mature GDF11 ligand is inhibited by its own prodomain, and extracellular proteolysis of this prodomain is thought to be necessary for GDF11 activity. The contribution of this proteolytic regulatory mechanism to Smad activation during embryogenesis in vivo, and to the development of posterior pattern, has not been characterized. We investigate here the role of Xenopus GDF11 in the activation of Smad2 during the development of tailbud-stage embryos, and the role of this activation in larval development. We also demonstrate that the activity of BMP-1/Tolloid-like proteases is necessary for the normal GDF11-dependent activation of Smad2 phosphorylation during post-gastrula development. These data demonstrate that GDF11 has a central role in the activation of Smad2 phosphorylation in tailbud stage Xenopus embryos, and provide the first evidence that BMP-1/Tolloid-mediated prodomain cleavage is important for activation of GDF11 in vivo.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Smad2/metabolismo , Cola (estructura animal)/embriología , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Xenopus/genética , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 1/metabolismo , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/genética , Clonación Molecular , Técnicas de Silenciamiento del Gen , Datos de Secuencia Molecular , Fenotipo , Fosforilación , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Proteína Smad2/genética , Xenopus/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
11.
Dev Biol ; 295(2): 730-42, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16684517

RESUMEN

Activin/nodal-like TGF-beta superfamily ligands signal through the type I receptors Alk4, Alk5, and Alk7, and are responsible for mediating a number of essential processes in development. SB-431542, a chemical inhibitor of activin/nodal signaling, acts by specifically interfering with type I receptors. Here, we use inhibitor-resistant mutant receptors to examine the efficacy and specificity of SB-431542 in Xenopus and zebrafish embryos. Treatment with SB-431542 eliminates Smad2 phosphorylation in vivo and generates a phenotype very similar to those observed in genetic mutants in the nodal signaling pathway. Inhibitor-resistant Alk4 efficiently rescues Smad2 signaling, developmental phenotype, and marker gene expression after inhibitor treatment. This system was used to examine type I receptor specificity for several activin/nodal ligands. We find that Alk4 can efficiently rescue signaling by a wide range of ligands, while Alk7 can only weakly rescue signaling by the same ligands. In whole embryos, nodal signaling during gastrulation can be rescued with Alk4, but not Alk7, while Alk5 can only mediate signaling by ligands expressed later in development. The combination of the ALK inhibitor SB-431542 with inhibitor-resistant ALKs provides a powerful set of tools for examining nodal/activin signaling during embryogenesis.


Asunto(s)
Receptores de Activinas/metabolismo , Inhibidores Enzimáticos/farmacología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I , Animales , Benzamidas/farmacología , Dioxoles/farmacología , Embrión no Mamífero , Desarrollo Embrionario , Ligandos , Proteína Nodal , Proteína Smad2/metabolismo , Proteínas de Xenopus , Proteínas de Pez Cebra
12.
Development ; 131(14): 3263-72, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15201219

RESUMEN

Translational control is an essential mechanism of gene control utilized throughout development, yet the molecular mechanisms underlying translational activation and repression are poorly understood. We have investigated the translational control of the C. elegans caudal homolog, pal-1, and found that GLD-1, a member of the evolutionarily conserved STAR/Maxi-KH domain family, acts through a minimal pal-1 3' UTR element to repress pal-1 translation in the distal germline. We also provide data suggesting that GLD-1 may repress pal-1 translation after initiation. Finally, we show that GLD-1 represses the distal germline expression of the KH domain protein MEX-3, which was previously shown to repress PAL-1 expression in the proximal germline and which appears specialized to control PAL-1 expression patterns in the embryo. Hence, GLD-1 mediates a developmental switch in the control of PAL-1 repression, allowing MEX-3 to accumulate and take over the task of PAL-1 repression in the proximal germline, where GLD-1 protein levels decline.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Biosíntesis de Proteínas , Transactivadores/fisiología , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Evolución Molecular , Genes de Helminto , Genes Reporteros , Proteínas Fluorescentes Verdes , Proteínas del Helminto/química , Proteínas de Homeodominio/metabolismo , Proteínas Luminiscentes/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Interferencia de ARN , ARN de Helminto/química , ARN de Helminto/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasas/metabolismo , Ribosomas/metabolismo , Fracciones Subcelulares/metabolismo , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA