Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 293(35): 13496-13508, 2018 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-30018136

RESUMO

Hedgehog (Hh) signaling plays a key role in the development and maintenance of animal tissues. This signaling is mediated by the atypical G protein-coupled receptor (GPCR) Smoothened (Smo). Smo activation leads to signaling through several well-characterized effectors to activate Hh target gene expression. Recent studies have implicated activation of the heterotrimeric G protein subunit Gαi and the subsequent decrease in cellular cAMP levels in promoting the Hh response in flies and mammals. Although Hh stimulation decreases cAMP levels in some insect cell lines, here using a bioluminescence resonance energy transfer (BRET)-based assay we found that this stimulation had no detectable effect in Drosophila S2-R+ cells. However, we observed an unexpected and significant Gαs-dependent increase in cAMP levels in response to strong Smo activation in Smo-transfected cells. This effect was mediated by Smo's broadly conserved core, and was specifically activated in response to phosphorylation of the Smo C-terminus by GPCR kinase 2 (Gprk2). Genetic analysis of heterotrimeric G protein function in the developing Drosophila wing revealed a positive role for cAMP in the endogenous Hh response. Specifically, we found that mutation or depletion of Gαs diminished low-threshold Hh responses in Drosophila, whereas depletion of Gαi potentiated them (in contrast to previous findings). Our analysis suggested that regulated cAMP production is important for controlling the sensitivity of cellular responses to Hh in Drosophila.


Assuntos
AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Receptor Smoothened/metabolismo , Animais , Fosforilação
2.
J Biol Chem ; 290(34): 20960-20971, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26170449

RESUMO

Protein kinases carry out important functions in cells both by phosphorylating substrates and by means of regulated non-catalytic activities. Such non-catalytic functions have been ascribed to many kinases, including some members of the Ste20 family. The Drosophila Ste20 kinase Slik phosphorylates and activates Moesin in developing epithelial tissues to promote epithelial tissue integrity. It also functions non-catalytically to promote epithelial cell proliferation and tissue growth. We carried out a structure-function analysis to determine how these two distinct activities of Slik are controlled. We find that the conserved C-terminal coiled-coil domain of Slik, which is necessary and sufficient for apical localization of the kinase in epithelial cells, is not required for Moesin phosphorylation but is critical for the growth-promoting function of Slik. Slik is auto- and trans-phosphorylated in vivo. Phosphorylation of at least two of three conserved sites in the activation segment is required for both efficient catalytic activity and non-catalytic signaling. Slik function is thus dependent upon proper localization of the kinase via the C-terminal coiled-coil domain and activation via activation segment phosphorylation, which enhances both phosphorylation of substrates like Moesin and engagement of effectors of its non-catalytic growth-promoting activity.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Proteínas Serina-Treonina Quinases/genética , Sequência de Aminoácidos , Animais , Biocatálise , Técnicas de Cultura de Células , Proliferação de Células , Sequência Conservada , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transgenes
3.
PLoS Genet ; 10(7): e1004399, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25009998

RESUMO

Hedgehog (Hh) signaling is essential for normal growth, patterning, and homeostasis of many tissues in diverse organisms, and is misregulated in a variety of diseases including cancer. Cytoplasmic Hedgehog signaling is activated by multisite phosphorylation of the seven-pass transmembrane protein Smoothened (Smo) in its cytoplasmic C-terminus. Aside from a short membrane-proximal stretch, the sequence of the C-terminus is highly divergent in different phyla, and the evidence suggests that the precise mechanism of Smo activation and transduction of the signal to downstream effectors also differs. To clarify the conserved role of G-protein-coupled receptor kinases (GRKs) in Smo regulation, we mapped four clusters of phosphorylation sites in the membrane-proximal C-terminus of Drosophila Smo that are phosphorylated by Gprk2, one of the two fly GRKs. Phosphorylation at these sites enhances Smo dimerization and increases but is not essential for Smo activity. Three of these clusters overlap with regulatory phosphorylation sites in mouse Smo and are highly conserved throughout the bilaterian lineages, suggesting that they serve a common function. Consistent with this, we find that a C-terminally truncated form of Drosophila Smo consisting of just the highly conserved core, including Gprk2 regulatory sites, can recruit the downstream effector Costal-2 and activate target gene expression, in a Gprk2-dependent manner. These results indicate that GRK phosphorylation in the membrane proximal C-terminus is an evolutionarily ancient mechanism of Smo regulation, and point to a higher degree of similarity in the regulation and signaling mechanisms of bilaterian Smo proteins than has previously been recognized.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Quinase 2 de Receptor Acoplado a Proteína G/genética , Regulação da Expressão Gênica no Desenvolvimento , Receptores Acoplados a Proteínas G/metabolismo , Animais , Proteínas de Drosophila/biossíntese , Drosophila melanogaster/crescimento & desenvolvimento , Quinase 2 de Receptor Acoplado a Proteína G/biossíntese , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Cinesinas/metabolismo , Camundongos , Fosforilação/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Receptor Smoothened
4.
Fly (Austin) ; 6(3): 135-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22653052

RESUMO

Hedgehog (Hh) signaling is essential for proper tissue patterning and maintenance and has a substantial impact on human disease. While many of the main components and mechanisms involved in transduction of the Hh signal have been identified, the details of how the pathway functions are continually being refined. One aspect that has attracted much attention recently is the involvement of G-protein-coupled receptor kinases (GRKs) in the pathway. These regulators of G-protein-coupled receptor (GPCR) signaling have an evolutionarily-conserved function in promoting high-threshold Hh target gene expression through regulation of Smoothened (Smo), a GPCR family member that activates intracellular Hh signaling. Several models of how GRKs impact on Smo to increase downstream signaling have been proposed. Recently, we demonstrated that these kinases have surprisingly complex and conflicting roles, acting to limit signaling through the pathway while also promoting Smo activity. In addition to the previously described direct effects of Gprk2 on Smo activation, Gprk2 also indirectly affects Hh signaling by controlling production of the second messenger cyclic AMP to influence Protein kinase A activity.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila/crescimento & desenvolvimento , Quinases de Receptores Acoplados a Proteína G/fisiologia , Proteínas Hedgehog/fisiologia , Modelos Biológicos , Animais , AMP Cíclico/genética , AMP Cíclico/metabolismo , AMP Cíclico/fisiologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Quinases de Receptores Acoplados a Proteína G/genética , Regulação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Transdução de Sinais
5.
Development ; 139(1): 85-94, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22096079

RESUMO

G-protein-coupled receptor kinases (GRKs) play a conserved role in Hedgehog (Hh) signaling. In several systems, GRKs are required for efficient Hh target gene expression. Their principal target appears to be Smoothened (Smo), the intracellular signal-generating component of the pathway and a member of the G-protein-coupled receptor (GPCR) protein family. In Drosophila, a GRK called Gprk2 is needed for internalization and downregulation of activated Smo, consistent with the typical role of these kinases in negatively regulating GPCRs. However, Hh target gene activation is strongly impaired in gprk2 mutant flies, indicating that Gprk2 must also positively regulate Hh signaling at some level. To investigate its function in signaling, we analyzed several different readouts of Hh pathway activity in animals or cells lacking Gprk2. Surprisingly, although target gene expression was impaired, Smo-dependent activation of downstream components of the signaling pathway was increased in the absence of Gprk2. This suggests that Gprk2 does indeed play a role in terminating Smo signaling. However, loss of Gprk2 resulted in a decrease in cellular cAMP concentrations to a level that was limiting for Hh target gene activation. Normal expression of target genes was restored in gprk2 mutants by stimulating cAMP production or activating the cAMP-dependent Protein kinase A (Pka). Our results suggest that direct regulation of Smo by Gprk2 is not absolutely required for Hh target gene expression. Gprk2 is important for normal cAMP regulation, and thus has an indirect effect on the activity of Pka-regulated components of the Hh pathway, including Smo itself.


Assuntos
AMP Cíclico/metabolismo , Drosophila melanogaster/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Ácidos Graxos Insaturados , Plasmídeos/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Ativação Transcricional/fisiologia
6.
Dev Biol ; 337(1): 99-109, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19850026

RESUMO

The Hedgehog (Hh) signaling pathway plays a conserved and essential role in regulating development and homeostasis of numerous tissues. Cytoplasmic signaling is initiated by Smoothened (Smo), a G-protein-coupled receptor (GPCR) family member, whose levels and activity are regulated by the Hh receptor Patched (Ptc). In response to Hh binding to Ptc, Ptc-mediated repression of Smo is relieved, leading to Smo activation, surface accumulation, and downstream signaling. We find that downregulation of Drosophila Smo protein in Hh-responding imaginal disc cells is dependent on the activity of G-protein-coupled receptor kinase 2 (Gprk2). By analyzing gain- and null loss-of-function phenotypes, we provide evidence that Gprk2 promotes Smo internalization subsequent to its activation, most likely by direct phosphorylation. Ptc-dependent regulation of Smo accumulation is normal in gprk2 mutants, indicating that Gprk2 and Ptc downregulate Smo by different mechanisms. Finally, we show that both Drosophila G-protein-coupled receptor kinase orthologues, Gprk1 and Gprk2, act in a partially redundant manner to promote Hh signaling. Our results suggest that Smo is regulated by distinct Ptc-dependent and Gprk2-dependent trafficking mechanisms in vivo, analogous to constitutive and activity-dependent regulation of GPCRs. G-protein-coupled receptor kinase activity is also important for efficient downstream signaling.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Regulação da Expressão Gênica , Receptores Acoplados a Proteínas G/genética , Animais , Arrestinas/fisiologia , Células Cultivadas , Drosophila melanogaster , Receptor Quinase 1 Acoplada a Proteína G/fisiologia , Proteínas Hedgehog/fisiologia , Fosforilação , Receptores de Superfície Celular/fisiologia , Transdução de Sinais , Receptor Smoothened , Temperatura , beta-Arrestinas
7.
Fungal Genet Biol ; 45(11): 1497-505, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18812228

RESUMO

Neurosporaxanthin, beta-apo-4'-carotenoic acid (C35), represents the end-product of the carotenoid pathway in Neurospora crassa. It is supposed to be synthesized in three steps catalyzed by sequential AL-2, CAO-2 and YLO-1 activities: (i) cyclization of 3,4-didehydrolycopene (C40); (ii) cleavage of torulene into beta-apo-4'-carotenal (C35); and finally (iii) oxidation of beta-apo-4'-carotenal. However, analyses of the ylo-1 mutant revealed the accumulation of intermediates other than beta-apo-4'-carotenal. Here, we generated a 3,4-didehydrolycopene accumulating Escherichia coli strain and showed that CAO-2 cleaves this acyclic carotene in vivo and in vitro yielding apo-4'-lycopenal. The apocarotenoids accumulated in the ylo-1 mutant were then identified as apo-4'-lycopenal and apo-4'-lycopenol, pointing to the former as the YLO-1 substrate and indicating that cyclization is the last step in neurosporaxanthin biosynthesis. This was further substantiated by analyses of a cyclase-deficient al-2 mutant, revealing the accumulation of apo-4'-lycopenoic acid. The three acyclic apocarotenoids presented here have not been found naturally before.


Assuntos
Vias Biossintéticas , Carotenoides/biossíntese , Mutação , Neurospora crassa/genética , Neurospora crassa/metabolismo , Carotenoides/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Lactonas/metabolismo , Neurospora crassa/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...