Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nature ; 533(7603): 411-5, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27193687

RESUMEN

Obesity and its associated comorbidities (for example, diabetes mellitus and hepatic steatosis) contribute to approximately 2.5 million deaths annually and are among the most prevalent and challenging conditions confronting the medical profession. Neurotensin (NT; also known as NTS), a 13-amino-acid peptide predominantly localized in specialized enteroendocrine cells of the small intestine and released by fat ingestion, facilitates fatty acid translocation in rat intestine, and stimulates the growth of various cancers. The effects of NT are mediated through three known NT receptors (NTR1, 2 and 3; also known as NTSR1, 2, and NTSR3, respectively). Increased fasting plasma levels of pro-NT (a stable NT precursor fragment produced in equimolar amounts relative to NT) are associated with increased risk of diabetes, cardiovascular disease and mortality; however, a role for NT as a causative factor in these diseases is unknown. Here we show that NT-deficient mice demonstrate significantly reduced intestinal fat absorption and are protected from obesity, hepatic steatosis and insulin resistance associated with high fat consumption. We further demonstrate that NT attenuates the activation of AMP-activated protein kinase (AMPK) and stimulates fatty acid absorption in mice and in cultured intestinal cells, and that this occurs through a mechanism involving NTR1 and NTR3 (also known as sortilin). Consistent with the findings in mice, expression of NT in Drosophila midgut enteroendocrine cells results in increased lipid accumulation in the midgut, fat body, and oenocytes (specialized hepatocyte-like cells) and decreased AMPK activation. Remarkably, in humans, we show that both obese and insulin-resistant subjects have elevated plasma concentrations of pro-NT, and in longitudinal studies among non-obese subjects, high levels of pro-NT denote a doubling of the risk of developing obesity later in life. Our findings directly link NT with increased fat absorption and obesity and suggest that NT may provide a prognostic marker of future obesity and a potential target for prevention and treatment.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Neurotensina/metabolismo , Obesidad/inducido químicamente , Obesidad/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Drosophila melanogaster/metabolismo , Células Enteroendocrinas/metabolismo , Activación Enzimática , Cuerpo Adiposo/metabolismo , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Femenino , Humanos , Resistencia a la Insulina/fisiología , Mucosa Intestinal/metabolismo , Intestinos/citología , Metabolismo de los Lípidos , Masculino , Ratones , Persona de Mediana Edad , Neurotensina/sangre , Neurotensina/deficiencia , Neurotensina/genética , Obesidad/sangre , Obesidad/prevención & control , Precursores de Proteínas/sangre , Precursores de Proteínas/metabolismo
2.
Dev Biol ; 457(1): 128-139, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31550483

RESUMEN

Hedgehog (Hh) signaling has been shown to regulate multiple developmental processes, however, it is unclear how it regulates lipid metabolism. Here, we demonstrate that Hh signaling exhibits potent activity in Drosophila fat body, which is induced by both locally expressed and midgut-derived Hh proteins. Inactivation of Hh signaling increases, whereas activation of Hh signaling decreases lipid accumulation. The major lipase Brummer (Bmm) acts downstream of Smoothened (Smo) in Hh signaling to promote lipolysis, therefore, the breakdown of triacylglycerol (TAG). We identify a critical Ci binding site in bmm promoter that is responsible to mediate Bmm expression induced by Hh signaling. Genomic mutation of the Ci binding site significantly reduces the expression of Bmm and dramatically decreases the responsiveness to Ci overexpression. Together, our findings provide a model for lipolysis to be regulated by Hh signaling, raising the possibility for Hh signaling to be involved in lipid metabolic and/or lipid storage diseases.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/metabolismo , Lipasa/genética , Lipólisis , Transducción de Señal , Adipocitos/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/metabolismo , Cuerpo Adiposo/metabolismo , Femenino , Proteínas Hedgehog/metabolismo , Larva/metabolismo , Masculino , Receptor Smoothened/metabolismo , Factores de Transcripción/metabolismo
3.
FASEB J ; 34(6): 8596-8610, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32359121

RESUMEN

We previously reported that high levels of plasma neurotensin (NT), a gut hormone released from enteroendocrine cells of the small bowel, contribute to obesity and comorbid conditions. Gut microbiota has been implicated in the obesity development. Paneth cells are critical in maintaining gut microbiota composition and homeostasis by releasing antimicrobial proteins including α-defensins. The purpose of our current study was to determine the possible role of NT in gut microbiota composition and α-defensin gene expression associated with obesity. Here we show that the ratio of Firmicutes/Bacteroidetes (F/B ratio) and intestinal proinflammatory cytokines is significantly increased in NT+/+ mice fed with a high-fat diet (HFD) which were improved in NT-deficient mice. HFD disrupted the intestinal Mmp7/α-defensin axis, which was completely prevented in NT-/- mice. In addition, NT treatment inhibited DEFA5 expression and concurrent NF-κB activity, which was blocked by a pan PKC inhibitor (Gö6983) or an inhibitor for atypical PKCs (CRT0066854). More importantly, the shRNA-mediated knockdown of atypical PKCτ reversed NT-attenuated DEFA5 expression and increased NF-κB activity. NT contributes to the HFD-induced disruption of gut microbiota composition and α-defensin expression. PKCτ/λ plays a central role in NT-mediated α-defensin gene expression which might be mediated through the inhibition of NF-κB signaling pathways in Paneth cells.


Asunto(s)
Disbiosis/metabolismo , Inflamación/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Neurotensina/metabolismo , alfa-Defensinas/metabolismo , Tejido Adiposo/metabolismo , Animales , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Disbiosis/patología , Microbioma Gastrointestinal/fisiología , Inflamación/patología , Resistencia a la Insulina/fisiología , Intestinos/patología , Masculino , Ratones , Ratones Obesos , FN-kappa B/metabolismo , Obesidad/metabolismo , Células de Paneth/metabolismo , Transducción de Señal/fisiología
4.
J Cell Sci ; 131(1)2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29142103

RESUMEN

Smoothened (Smo), a GPCR family protein, plays a critical role in the reception and transduction of Hedgehog (Hh) signal. Smo is phosphorylated and activated on the cell surface; however, it is unknown whether Smo can be intracellularly activated. Here, we demonstrate that inactivation of the ESCRT-III causes dramatic accumulation of Smo in the ESCRT-III/MVB compartment, and subsequent activation of Hh signaling. In contrast, inactivation of ESCRTs 0-II induces mild Smo accumulation in the ESCRT-III/MVB compartment. We provide evidence that Kurtz (Krz), the Drosophila ß-arrestin2, acts in parallel with the ESCRTs 0-II pathway to sort Smo to the multivesicular bodies and lysosome-mediated degradation. Additionally, upon inactivation of ESCRT-III, all active and inactive forms of Smo are accumulated. Endogenous Smo accumulated upon ESCRT-III inactivation is highly activated, which is induced by phosphorylation but not sumoylation. Taken together, our findings demonstrate a model for intracellular activation of Smo, raising the possibility for tissue overgrowth caused by an excessive amount, rather than mutation of Smo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Animales , Arrestinas/genética , Arrestinas/metabolismo , Membrana Celular/metabolismo , Drosophila/citología , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Fosforilación , Transporte de Proteínas/genética , Receptor Smoothened/genética , Sumoilación
5.
PLoS Biol ; 14(2): e1002375, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26863604

RESUMEN

In Hedgehog (Hh) signaling, binding of Hh to the Patched-Interference Hh (Ptc-Ihog) receptor complex relieves Ptc inhibition on Smoothened (Smo). A longstanding question is how Ptc inhibits Smo and how such inhibition is relieved by Hh stimulation. In this study, we found that Hh elevates production of phosphatidylinositol 4-phosphate (PI(4)P). Increased levels of PI(4)P promote, whereas decreased levels of PI(4)P inhibit, Hh signaling activity. We further found that PI(4)P directly binds Smo through an arginine motif, which then triggers Smo phosphorylation and activation. Moreover, we identified the pleckstrin homology (PH) domain of G protein-coupled receptor kinase 2 (Gprk2) as an essential component for enriching PI(4)P and facilitating Smo activation. PI(4)P also binds mouse Smo (mSmo) and promotes its phosphorylation and ciliary accumulation. Finally, Hh treatment increases the interaction between Smo and PI(4)P but decreases the interaction between Ptc and PI(4)P, indicating that, in addition to promoting PI(4)P production, Hh regulates the pool of PI(4)P associated with Ptc and Smo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas Hedgehog/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Cilios/metabolismo , Drosophila , Ratones , Células 3T3 NIH , Receptores Patched , Receptor Patched-1 , Fosforilación , Receptores de Superficie Celular/metabolismo , Receptor Smoothened
6.
PLoS Genet ; 12(5): e1006054, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27195754

RESUMEN

Hedgehog (Hh) signaling regulates multiple aspects of metazoan development and tissue homeostasis, and is constitutively active in numerous cancers. We identified Ubr3, an E3 ubiquitin ligase, as a novel, positive regulator of Hh signaling in Drosophila and vertebrates. Hh signaling regulates the Ubr3-mediated poly-ubiquitination and degradation of Cos2, a central component of Hh signaling. In developing Drosophila eye discs, loss of ubr3 leads to a delayed differentiation of photoreceptors and a reduction in Hh signaling. In zebrafish, loss of Ubr3 causes a decrease in Shh signaling in the developing eyes, somites, and sensory neurons. However, not all tissues that require Hh signaling are affected in zebrafish. Mouse UBR3 poly-ubiquitinates Kif7, the mammalian homologue of Cos2. Finally, loss of UBR3 up-regulates Kif7 protein levels and decreases Hh signaling in cultured cells. In summary, our work identifies Ubr3 as a novel, evolutionarily conserved modulator of Hh signaling that boosts Hh in some tissues.


Asunto(s)
Proteínas de Drosophila/genética , Ojo/metabolismo , Cinesinas/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Ojo/crecimiento & desarrollo , Proteínas Hedgehog/genética , Cinesinas/metabolismo , Ratones , Células Fotorreceptoras/metabolismo , Poliubiquitina , Proteolisis , ARN Interferente Pequeño , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Pez Cebra/genética
7.
Genes Dev ; 24(18): 2054-67, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20844016

RESUMEN

G protein-coupled receptor kinase 2 (Gprk2/GRK2) plays a conserved role in modulating Hedgehog (Hh) pathway activity, but its mechanism of action remains unknown. Here we provide evidence that Gprk2 promotes high-level Hh signaling by regulating Smoothened (Smo) conformation through both kinase-dependent and kinase-independent mechanisms. Gprk2 promotes Smo activation by phosphorylating Smo C-terminal tail (C-tail) at Ser741/Thr742, which is facilitated by PKA and CK1 phosphorylation at adjacent Ser residues. In addition, Gprk2 forms a dimer/oligomer and binds Smo C-tail in a kinase activity-independent manner to stabilize the active Smo conformation, and promotes dimerization/oligomerization of Smo C-tail. Gprk2 expression is induced by Hh signaling, and Gprk2/Smo interaction is facilitated by PKA/CK1-mediated phosphorylation of Smo C-tail. Thus, Gprk2 forms a positive feedback loop and acts downstream from PKA and CK1 to facilitate high-level Hh signaling by promoting the active state of Smo through direct phosphorylation and molecular scaffolding.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Drosophila/enzimología , Proteínas de Drosophila/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Proteínas Hedgehog/genética , Fosforilación , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened
8.
PLoS Pathog ; 11(3): e1004783, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25815785

RESUMEN

Stimulator of interferon genes (STING, also known as MITA and ERIS) is critical in protecting the host against DNA pathogen invasion. However, the molecular mechanism underlying the regulation of STING remains unclear. Here, we show that PPM1A negatively regulates antiviral signaling by targeting STING in its phosphatase activity-dependent manner, and in a line with this, PPM1A catalytically dephosphorylates STING and TBK1 in vitro. Importantly, we provide evidence that whereas TBK1 promotes STING aggregation in a phosphorylation-dependent manner, PPM1A antagonizes STING aggregation by dephosphorylating both STING and TBK1, emphasizing that phosphorylation is crucial for the efficient activation of STING. Our findings demonstrate a novel regulatory circuit in which STING and TBK1 reciprocally regulate each other to enable efficient antiviral signaling activation, and PPM1A dephosphorylates STING and TBK1, thereby balancing this antiviral signal transduction.


Asunto(s)
Proteínas de la Membrana/inmunología , Fosfoproteínas Fosfatasas/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal/inmunología , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Fosfoproteínas Fosfatasas/genética , Fosforilación/genética , Fosforilación/inmunología , Proteína Fosfatasa 2C , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Células Vero
9.
Proc Natl Acad Sci U S A ; 111(45): E4842-50, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349414

RESUMEN

Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Cell surface/cilium accumulation of Smo is thought to play an important role in Hh signaling, but how the localization of Smo is controlled remains poorly understood. In this study, we demonstrate that atypical PKC (aPKC) regulates Smo phosphorylation and basolateral accumulation in Drosophila wings. Inactivation of aPKC by either RNAi or a mutation inhibits Smo basolateral accumulation and attenuates Hh target gene expression. In contrast, expression of constitutively active aPKC elevates basolateral accumulation of Smo and promotes Hh signaling. The aPKC-mediated phosphorylation of Smo at Ser680 promotes Ser683 phosphorylation by casein kinase 1 (CK1), and these phosphorylation events elevate Smo activity in vivo. Moreover, aPKC has an additional positive role in Hh signaling by regulating the activity of Cubitus interruptus (Ci) through phosphorylation of the Zn finger DNA-binding domain. Finally, the expression of aPKC is up-regulated by Hh signaling in a Ci-dependent manner. Our findings indicate a direct involvement of aPKC in Hh signaling beyond its role in cell polarity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas Hedgehog/metabolismo , Proteína Quinasa C/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas Hedgehog/genética , Fosforilación/fisiología , Proteína Quinasa C/genética , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened , Factores de Transcripción/genética , Regulación hacia Arriba/fisiología , Alas de Animales/metabolismo
10.
PLoS Biol ; 10(1): e1001238, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22253573

RESUMEN

The seven transmembrane protein Smoothened (Smo) is a critical component of the Hedgehog (Hh) signaling pathway and is regulated by phosphorylation, dimerization, and cell-surface accumulation upon Hh stimulation. However, it is not clear how Hh regulates Smo accumulation on the cell surface or how Hh regulates the intracellular trafficking of Smo. In addition, little is known about whether ubiquitination is involved in Smo regulation. In this study, we demonstrate that Smo is multi-monoubiquitinated and that Smo ubiquitination is inhibited by Hh and by phosphorylation. Using an in vivo RNAi screen, we identified ubiquitin-specific protease 8 (USP8) as a deubiquitinase that down-regulates Smo ubiquitination. Inactivation of USP8 increases Smo ubiquitination and attenuates Hh-induced Smo accumulation, leading to decreased Hh signaling activity. Moreover, overexpression of USP8 prevents Smo ubiquitination and elevates Smo accumulation, leading to increased Hh signaling activity. Mechanistically, we show that Hh promotes the interaction of USP8 with Smo aa625-753, which covers the three PKA and CK1 phosphorylation clusters. Finally, USP8 promotes the accumulation of Smo at the cell surface and prevents localization to the early endosomes, presumably by deubiquitinating Smo. Our studies identify USP8 as a positive regulator in Hh signaling by down-regulating Smo ubiquitination and thereby mediating Smo intracellular trafficking.


Asunto(s)
Endopeptidasas/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Ubiquitina Tiolesterasa/fisiología , Animales , Drosophila , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Células 3T3 NIH , Transporte de Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Receptor Smoothened , Fracciones Subcelulares/metabolismo , Distribución Tisular , Transfección , Ubiquitina Tiolesterasa/genética , Ubiquitinación/genética
11.
J Biol Chem ; 288(45): 32809-32820, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24072710

RESUMEN

The Hedgehog (Hh) pathway is essential for embryonic development and adult tissue homeostasis. The Gli/Cubitus interruptus (Ci) family of transcription factors acts at the downstream end of the pathway to mediate Hh signaling. Both Hh-dependent and -independent Gli regulatory mechanisms are important for the output of Hh signaling. Daz interacting protein 1 (Dzip1) has bipartite positive and negative functions in the Hh pathway. The positive Hh regulatory function appears to be attributed to a requirement for Dzip1 during ciliogenesis. The mechanism by which Dzip1 inhibits Hh signaling, however, remains largely unclear. We recently found that Dzip1 is required for Gli turnover, which may account for its inhibitory function in Hh signaling. Here, we report that Dzip1 regulates Gli/Ci turnover by preventing degradation of speckle-type POZ protein (Spop), a protein that promotes proteasome-dependent turnover of Gli proteins. We provide evidence that Dzip1 regulates the stability of Spop independent of its function in ciliogenesis. Partial knockdown of Dzip1 to levels insufficient for perturbing ciliogenesis, sensitized Xenopus embryos to Hh signaling, leading to phenotypes that resemble activation of Hh signaling. Importantly, overexpression of Spop was able to restore proper Gli protein turnover and rescue phenotypes in Dzip1-depleted embryos. Consistently, depletion of Dzip1 in Drosophila S2 cells destabilized Hh-induced BTB protein (HIB), the Drosophila homolog of Spop, and increased the level of Ci. Thus, Dzip1-dependent stabilization of Spop/HIB is evolutionarily conserved and essential for proper regulation of Gli/Ci proteins in the Hh pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Cilios/genética , Cilios/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Embrión no Mamífero/embriología , Proteínas Hedgehog/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Estabilidad Proteica , Proteínas Represoras/genética , Factores de Transcripción/genética , Complejos de Ubiquitina-Proteína Ligasa , Proteínas de Xenopus/genética , Xenopus laevis , Proteína con Dedos de Zinc GLI1
12.
Development ; 138(19): 4219-31, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21852395

RESUMEN

Hedgehog (Hh) transduces signal by regulating the subcellular localization and conformational state of the GPCR-like protein Smoothened (Smo) but how Smo relays the signal to cytoplasmic signaling components remains poorly understood. Here, we show that Hh-induced Smo conformational change recruits Costal2 (Cos2)/Fused (Fu) and promotes Fu kinase domain dimerization. We find that induced dimerization through the Fu kinase domain activates Fu by inducing multi-site phosphorylation of its activation loop (AL) and phospho-mimetic mutations of AL activate the Hh pathway. Interestingly, we observe that graded Hh signals progressively increase Fu kinase domain dimerization and AL phosphorylation, suggesting that Hh activates Fu in a dose-dependent manner. Moreover, we find that activated Fu regulates Cubitus interruptus (Ci) by both promoting its transcriptional activator activity and inhibiting its proteolysis into a repressor form. We provide evidence that activated Fu exerts these regulations by interfering with the formation of Ci-Sufu and Ci-Cos2-kinase complexes that normally inhibit Ci activity and promote its processing. Taken together, our results suggest that Hh-induced Smo conformational change facilitates the assembly of active Smo-Cos2-Fu signaling complexes that promote Fu kinase domain dimerization, phosphorylation and activation, and that Fu regulates both the activator and repressor forms of Ci.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Cinesinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Dimerización , Drosophila melanogaster/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Fosforilación , Unión Proteica , Conformación Proteica , Interferencia de ARN , Transducción de Señal , Receptor Smoothened
13.
PLoS Biol ; 9(6): e1001083, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21695114

RESUMEN

Hedgehog (Hh) signaling regulates embryonic development and adult tissue homeostasis through the GPCR-like protein Smoothened (Smo), but how vertebrate Smo is activated remains poorly understood. In Drosophila, Hh dependent phosphorylation activates Smo. Whether this is also the case in vertebrates is unclear, owing to the marked sequence divergence between vertebrate and Drosophila Smo (dSmo) and the involvement of primary cilia in vertebrate Hh signaling. Here we demonstrate that mammalian Smo (mSmo) is activated through multi-site phosphorylation of its carboxyl-terminal tail by CK1α and GRK2. Phosphorylation of mSmo induces its active conformation and simultaneously promotes its ciliary accumulation. We demonstrate that graded Hh signals induce increasing levels of mSmo phosphorylation that fine-tune its ciliary localization, conformation, and activity. We show that mSmo phosphorylation is induced by its agonists and oncogenic mutations but is blocked by its antagonist cyclopamine, and efficient mSmo phosphorylation depends on the kinesin-II ciliary motor. Furthermore, we provide evidence that Hh signaling recruits CK1α to initiate mSmo phosphorylation, and phosphorylation further increases the binding of CK1α and GRK2 to mSmo, forming a positive feedback loop that amplifies and/or sustains mSmo phosphorylation. Hence, despite divergence in their primary sequences and their subcellular trafficking, mSmo and dSmo employ analogous mechanisms for their activation.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Cilios/metabolismo , Drosophila/genética , Drosophila/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , Transducción de Señal , Receptor Smoothened , Transfección
14.
Dev Biol ; 366(2): 172-84, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22537496

RESUMEN

The activation of Smoothened (Smo) requires phosphorylation at three clusters of Serine residues in Drosophila Hedgehog (Hh) signaling. However, the mechanism by which phosphorylation promotes Smo conformational change and subsequently activates Smo in response to Hh gradient remains unclear. Here, we show that the conformational states of Smo are determined by not only the amount but also the position of the negative charges provided by phosphorylation. By using a Smo phospho-specific antibody, we demonstrate that Smo is differentially phosphorylated at three clusters of serine residues in response to levels of Hh activity. Mutating the first cluster, compared to mutating the other clusters, impairs Smo activity more severely, whereas mutating the last cluster prohibits C-terminus dimerization. In addition, phosphorylation of the membrane proximal cluster promotes phosphorylation of the distal cluster. We propose a zipper-lock model in which the gradual phosphorylation at these clusters induces a gradual conformational change in the Smo cytoplasmic tail, which promotes the interaction between Smo and Costal2 (Cos2). Moreover, we show that Hh regulates both PKA and CK1 phosphorylation of Smo. Thus, the differential phosphorylation of Smo mediates the thresholds of Hh activity.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proteínas de Drosophila/química , Modelos Moleculares , Fosforilación , Conformación Proteica , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/química , Transducción de Señal , Receptor Smoothened
15.
Sci Signal ; 16(807): eadd6834, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37847757

RESUMEN

Hedgehog (Hh) signaling controls growth and patterning during embryonic development and homeostasis in adult tissues. Hh binding to the receptor Patched (Ptc) elicits intracellular signaling by relieving Ptc-mediated inhibition of the transmembrane protein Smoothened (Smo). We uncovered a role for the lipid phosphatidic acid (PA) in the regulation of the Hh pathway in Drosophila melanogaster. Deleting the Ptc C-terminal tail or mutating the predicted PA-binding sites within it prevented Ptc from inhibiting Smo in wing discs and in cultured cells. The C-terminal tail of Ptc directly interacted with PA in vitro, an association that was reduced by Hh, and increased the amount of PA at the plasma membrane in cultured cells. Smo also interacted with PA in vitro through a binding pocket located in the transmembrane region, and mutating residues in this pocket reduced Smo activity in vivo and in cells. By genetically manipulating PA amounts in vivo or treating cultured cells with PA, we demonstrated that PA promoted Smo activation. Our findings suggest that Ptc may sequester PA in the absence of Hh and release it in the presence of Hh, thereby increasing the amount of PA that is locally available to promote Smo activation.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened/genética , Receptor Smoothened/metabolismo
16.
J Biol Chem ; 286(42): 36171-9, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21878643

RESUMEN

The Hedgehog (Hh) pathway is evolutionarily conserved and plays critical roles during embryonic development and adult tissue homeostasis. Defective Hh signaling has been linked to a wide range of birth defects and cancers. Hh family proteins regulate the expression of their downstream target genes through the control of proteolytic processing and the transcriptional activation function of Gli transcription factors. Although Hh-dependent regulation of Gli has been studied extensively, other Gli regulatory mechanisms remain relatively unappreciated. Here we report our identification of a novel signaling cascade that controls the stability of Gli proteins. This cascade consists of Daz interacting protein 1 (Dzip1), casein kinase 2 (CK2), and B56 containing protein phosphatase 2As (PP2As). We provide evidence that Dzip1 is involved in a novel Gli turnover pathway. We show that CK2 directly phosphorylates Dzip1 at four serine residues, Ser-664/665/706/714. B56-containing PP2As, through binding to a domain located between amino acid residue 474 and 550 of Dzip1, dephosphorylate Dzip1 on these CK2 sites. Our mutagenesis analysis further demonstrates that the unphosphorylatable form of Dzip1 is more potent in promoting Gli turnover. Consistently, we found that the stability of Gli proteins was decreased upon CK2 inhibition and increased by inhibition of B56-containing PP2As. Thus, reversible phosphorylation of Dzip1, which is controlled by the antagonistic action of CK2 and B56-containing PP2As, has an important impact on the stability of Gli transcription factors and Hh signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa de la Caseína II/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína Fosfatasa 2/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Quinasa de la Caseína II/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Mutagénesis , Células 3T3 NIH , Fosforilación/fisiología , Proteína Fosfatasa 2/genética , Estabilidad Proteica , Estructura Terciaria de Proteína , Transducción de Señal/fisiología , Factores de Transcripción/genética , Xenopus laevis , Proteína con Dedos de Zinc GLI1
17.
Front Cell Dev Biol ; 10: 847844, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35321245

RESUMEN

The GPCR-family protein Smoothened (Smo) is essential for Hedgehog (Hh) signal transduction in both insects and vertebrates. The regulation of subcellular localization and abundance of Smo is a critical step in Hh signaling. Recent studies have demonstrated that Smo is subjected to ubiquitination mediated by multiple E3 ubiquitin ligases, leading to Smo endocytosis and subsequent degradation through the proteasome- and lysosome-mediated pathways in Drosophila. Ubiquitination of Smo also promotes its ciliary exit in mammalian cells. Hh inhibits Smo ubiquitination by blocking E3 ligase recruitment and promoting Smo deubiquitination through the ubiquitin-specific protease 8 (USP8) in Drosophila. Inhibition of Smo ubiquitination by Hh promotes Smo cell surface accumulation in Drosophila and ciliary accumulation in mammalian cells. Interestingly, Hh also induces sumoylation of Smo in both Drosophila and mammalian cells, which promotes Smo cell surface/ciliary accumulation. This review focuses on how ubiquitination and sumoylation regulate Smo intracellular trafficking and abundance and how these processes are regulated by Hh.

18.
Methods Mol Biol ; 2374: 121-137, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34562248

RESUMEN

The GPCR-family protein Smoothened (Smo) is an obligatory signal transducer of the Hedgehog (Hh) signaling pathway. Binding of Hh to its receptor Patched (Ptc) alleviates Ptc-mediated inhibition of Smo, allowing Smo to activate the Cubitus interruptus (Ci)/Gli family of zinc finger transcription factors. The activation of Smo is an early and crucial event in Hh signal transduction. Studies have shown that Hh induces cell surface/ciliary accumulation and phosphorylation of Smo by multiple kinases, including protein kinase A (PKA), casein kinase 1 (CK1), casein kinase 2 (CK2), G protein-coupled receptor kinase 2 (Gprk2/GRK2), and atypical PKC (aPKC). Here, we describe the assays used to examine the phosphorylation and activity of Smo, including in vitro kinase assay, phospho-specific antibodies, luciferase reporter assay, cell surface accumulation, and ciliary localization assays. These assays provide powerful tools to study Smo phosphorylation and activation, leading to mechanistic insight into Smo regulation.


Asunto(s)
Receptor Smoothened/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Fosforilación , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened/genética , Factores de Transcripción/metabolismo
19.
Cell Mol Gastroenterol Hepatol ; 13(2): 501-516, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34560309

RESUMEN

BACKGROUND & AIMS: Intestinal stem cells (ISCs) are sensitive to dietary alterations and nutrient availability. Neurotensin (NT), a gut peptide localized predominantly to the small bowel and released by fat ingestion, stimulates the growth of intestinal mucosa under basal conditions and during periods of nutrient deprivation, suggesting a possible role for NT on ISC function. METHODS: Leucine-rich repeat-containing G-protein coupled receptor 5-Enhanced Green Fluorescent Protein (Lgr5-EGFP) NT wild type (Nt+/+) and Lgr5-EGFP NT knockout (Nt-/-) mice were fed ad libitum or fasted for 48 hours. Small intestine tissue and crypts were examined by gene expression analyses, fluorescence-activated cell sorting, Western blot, immunohistochemistry, and crypt-derived organoid culture. Drosophila expressing NT in midgut enteroendocrine cells were fed a standard diet or low-energy diet and esg-green fluorescent protein+ ISCs were quantified via immunofluorescence. RESULTS: Loss of NT impaired crypt cell proliferation and ISC function in a manner dependent on nutrient status. Under nutrient-rich conditions, NT stimulated extracellular signal-regulated kinases 1 and 2 signaling and the expression of genes that promote cell-cycle progression, leading to crypt cell proliferation. Under conditions of nutrient depletion, NT stimulated WNT/ß-catenin signaling and promoted an ISC gene signature, leading to enhanced ISC function. NT was required for the induction of WNT/ß-catenin signaling and ISC-specific gene expression during nutrient depletion, and loss of NT reduced crypt cell proliferation and impaired ISC function and Lgr5 expression in the intestine during fasting. Conversely, the expression of NT in midgut enteroendocrine cells of Drosophila prevented loss of ISCs during nutrient depletion. CONCLUSIONS: Collectively, our findings establish an evolutionarily conserved role for NT in ISC maintenance during nutritional stress. GSE182828.


Asunto(s)
Neurotensina , Células Madre , Animales , Proliferación Celular , Intestino Delgado , Ratones , Neurotensina/metabolismo , Nutrientes , Células Madre/metabolismo
20.
J Biol Chem ; 285(48): 37218-26, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20876583

RESUMEN

Casein kinase 2 (CK2) is a typical serine/threonine kinase consisting of α and ß subunits and has been implicated in many cellular and developmental processes. In this study, we demonstrate that CK2 is a positive regulator of the Hedgehog (Hh) signal transduction pathway. We found that inactivation of CK2 by CK2ß RNAi enhances the loss-of-Hh wing phenotype induced by a dominant negative form of Smoothened (Smo). CK2ß RNAi attenuates Hh-induced Smo accumulation and down-regulates Hh target gene expression, whereas increasing CK2 activity by coexpressing CK2α and CK2ß increases Smo accumulation and induces ectopic Hh target gene expression. We identified the serine residues in Smo that can be phosphorylated by CK2 in vitro. Mutating these serine residues attenuates the ability of Smo to transduce high level Hh signaling activity in vivo. Furthermore, we found that CK2 plays an additional positive role downstream of Smo by regulating the stability of full-length Cubitus interruptus (Ci). CK2ß RNAi promotes Ci degradation whereas coexpressing CK2α and CK2ß increases the half-life of Ci. We showed that CK2 prevents Ci ubiquitination and degradation by the proteasome. Thus, CK2 promotes Hh signaling activity by regulating multiple pathway components.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Quinasa de la Caseína II/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/química , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Proteínas Hedgehog/genética , Estabilidad Proteica , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened , Factores de Transcripción/química , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda