Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 92: 273-298, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37001135

RESUMEN

Ligands of the Hedgehog (HH) pathway are paracrine signaling molecules that coordinate tissue development in metazoans. A remarkable feature of HH signaling is the repeated use of cholesterol in steps spanning ligand biogenesis, secretion, dispersal, and reception on target cells. A cholesterol molecule covalently attached to HH ligands is used as a molecular baton by transfer proteins to guide their secretion, spread, and reception. On target cells, a signaling circuit composed of a cholesterol transporter and sensor regulates transmission of HH signals across the plasma membrane to the cytoplasm. The repeated use of cholesterol in signaling supports the view that the HH pathway likely evolved by coopting ancient systems to regulate the abundance or organization of sterol-like lipids in membranes.


Asunto(s)
Colesterol , Proteínas Hedgehog , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ligandos , Colesterol/metabolismo , Transducción de Señal , Esteroles/metabolismo
2.
Cell ; 168(1-2): 264-279.e15, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28086093

RESUMEN

The life cycle of a primary cilium begins in quiescence and ends prior to mitosis. In quiescent cells, the primary cilium insulates itself from contiguous dynamic membrane processes on the cell surface to function as a stable signaling apparatus. Here, we demonstrate that basal restriction of ciliary structure dynamics is established by the cilia-enriched phosphoinositide 5-phosphatase, Inpp5e. Growth induction displaces ciliary Inpp5e and accumulates phosphatidylinositol 4,5-bisphosphate in distal cilia. This change triggers otherwise-forbidden actin polymerization in primary cilia, which excises cilia tips in a process we call cilia decapitation. While cilia disassembly is traditionally thought to occur solely through resorption, we show that an acute loss of IFT-B through cilia decapitation precedes resorption. Finally, we propose that cilia decapitation induces mitogenic signaling and constitutes a molecular link between the cilia life cycle and cell-division cycle. This newly defined ciliary mechanism may find significance in cell proliferation control during normal development and cancer.


Asunto(s)
Ciclo Celular , Cilios/metabolismo , Actinas/metabolismo , Animales , Riñón/citología , Riñón/metabolismo , Ratones , Células 3T3 NIH , Fosfatidilinositol 4,5-Difosfato , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo
4.
Proc Natl Acad Sci U S A ; 121(7): e2318024121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38330014

RESUMEN

Lipid synthesis is regulated by the actions of Scap, a polytopic membrane protein that binds cholesterol in membranes of the endoplasmic reticulum (ER). When ER cholesterol levels are low, Scap activates SREBPs, transcription factors that upregulate genes for synthesis of cholesterol, fatty acids, and triglycerides. When ER cholesterol levels rise, the sterol binds to Scap, triggering conformational changes that prevent activation of SREBPs and halting synthesis of lipids. To achieve a molecular understanding of how cholesterol regulates the Scap/SREBP machine and to identify therapeutics for dysregulated lipid metabolism, cholesterol-mimetic compounds that specifically bind and inhibit Scap are needed. To accomplish this goal, we focused on Anthrolysin O (ALO), a pore-forming bacterial toxin that binds cholesterol with a specificity and sensitivity that is uncannily similar to Scap. We reasoned that a small molecule that would bind and inhibit ALO might also inhibit Scap. High-throughput screening of a ~300,000-compound library for ALO-binding unearthed one molecule, termed UT-59, which binds to Scap's cholesterol-binding site. Upon binding, UT-59 triggers the same conformation changes in Scap as those induced by cholesterol and blocks activation of SREBPs and lipogenesis in cultured cells. UT-59 also inhibits SREBP activation in the mouse liver. Unlike five previously reported inhibitors of SREBP activation, UT-59 is the only one that acts specifically by binding to Scap's cholesterol-binding site. Our approach to identify specific Scap inhibitors such as UT-59 holds great promise in developing therapeutic leads for human diseases stemming from elevated SREBP activation, such as fatty liver and certain cancers.


Asunto(s)
Toxinas Bacterianas , Lipogénesis , Animales , Ratones , Humanos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Colesterol/metabolismo , Toxinas Bacterianas/metabolismo
5.
Development ; 148(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486668

RESUMEN

Birth defects result from interactions between genetic and environmental factors, but the mechanisms remain poorly understood. We find that mutations and teratogens interact in predictable ways to cause birth defects by changing target cell sensitivity to Hedgehog (Hh) ligands. These interactions converge on a membrane protein complex, the MMM complex, that promotes degradation of the Hh transducer Smoothened (SMO). Deficiency of the MMM component MOSMO results in elevated SMO and increased Hh signaling, causing multiple birth defects. In utero exposure to a teratogen that directly inhibits SMO reduces the penetrance and expressivity of birth defects in Mosmo-/- embryos. Additionally, tissues that develop normally in Mosmo-/- embryos are refractory to the teratogen. Thus, changes in the abundance of the protein target of a teratogen can change birth defect outcomes by quantitative shifts in Hh signaling. Consequently, small molecules that re-calibrate signaling strength could be harnessed to rescue structural birth defects.


Asunto(s)
Anomalías Inducidas por Medicamentos/genética , Interacción Gen-Ambiente , Proteínas Hedgehog/metabolismo , Penetrancia , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Transducción de Señal , Receptor Smoothened/genética , Receptor Smoothened/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(11): 5883-5894, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32132204

RESUMEN

Liquid-liquid phase separation (LLPS) is involved in the formation of membraneless organelles (MLOs) associated with RNA processing. The RNA-binding protein TDP-43 is present in several MLOs, undergoes LLPS, and has been linked to the pathogenesis of amyotrophic lateral sclerosis (ALS). While some ALS-associated mutations in TDP-43 disrupt self-interaction and function, here we show that designed single mutations can enhance TDP-43 assembly and function via modulating helical structure. Using molecular simulation and NMR spectroscopy, we observe large structural changes upon dimerization of TDP-43. Two conserved glycine residues (G335 and G338) are potent inhibitors of helical extension and helix-helix interaction, which are removed in part by variants at these positions, including the ALS-associated G335D. Substitution to helix-enhancing alanine at either of these positions dramatically enhances phase separation in vitro and decreases fluidity of phase-separated TDP-43 reporter compartments in cells. Furthermore, G335A increases TDP-43 splicing function in a minigene assay. Therefore, the TDP-43 helical region serves as a short but uniquely tunable module where application of biophysical principles can precisely control assembly and function in cellular and synthetic biology applications of LLPS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Conformación Proteica en Hélice alfa , Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/genética , Escherichia coli/genética , Humanos , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Mutación , Conformación Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas , Empalme de Proteína , Proteínas de Unión al ARN/metabolismo
7.
EMBO J ; 37(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29438978

RESUMEN

TDP-43 is an RNA-binding protein active in splicing that concentrates into membraneless ribonucleoprotein granules and forms aggregates in amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. Although best known for its predominantly disordered C-terminal domain which mediates ALS inclusions, TDP-43 has a globular N-terminal domain (NTD). Here, we show that TDP-43 NTD assembles into head-to-tail linear chains and that phosphomimetic substitution at S48 disrupts TDP-43 polymeric assembly, discourages liquid-liquid phase separation (LLPS) in vitro, fluidizes liquid-liquid phase separated nuclear TDP-43 reporter constructs in cells, and disrupts RNA splicing activity. Finally, we present the solution NMR structure of a head-to-tail NTD dimer comprised of two engineered variants that allow saturation of the native polymerization interface while disrupting higher-order polymerization. These data provide structural detail for the established mechanistic role of the well-folded TDP-43 NTD in splicing and link this function to LLPS. In addition, the fusion-tag solubilized, recombinant form of TDP-43 full-length protein developed here will enable future phase separation and in vitro biochemical assays on TDP-43 function and interactions that have been hampered in the past by TDP-43 aggregation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Agregación Patológica de Proteínas/genética , Dominios Proteicos/genética , Empalme del ARN/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Polimerizacion , Polímeros/metabolismo , Agregación Patológica de Proteínas/patología
8.
Development ; 146(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092502

RESUMEN

Signaling pathways that mediate cell-cell communication are essential for collective cell behaviors in multicellular systems. The hedgehog (HH) pathway, first discovered and elucidated in Drosophila, is one of these iconic signaling systems that plays many roles during embryogenesis and in adults; abnormal HH signaling can lead to birth defects and cancer. We review recent structural and biochemical studies that have advanced our understanding of the vertebrate HH pathway, focusing on the mechanisms by which the HH signal is received by patched on target cells, transduced across the cell membrane by smoothened, and transmitted to the nucleus by GLI proteins to influence gene-expression programs.


Asunto(s)
Proteínas Hedgehog/metabolismo , Vertebrados/metabolismo , Animales , Proteínas Hedgehog/genética , Humanos , Transducción de Señal/genética , Transducción de Señal/fisiología
9.
Nat Chem Biol ; 16(12): 1303-1313, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33199907

RESUMEN

The Hedgehog (Hh) signaling pathway coordinates cell-cell communication in development and regeneration. Defects in this pathway underlie diseases ranging from birth defects to cancer. Hh signals are transmitted across the plasma membrane by two proteins, Patched 1 (PTCH1) and Smoothened (SMO). PTCH1, a transporter-like tumor-suppressor protein, binds to Hh ligands, but SMO, a G-protein-coupled-receptor family oncoprotein, transmits the Hh signal across the membrane. Recent structural, biochemical and cell-biological studies have converged at the surprising model that a specific pool of plasma membrane cholesterol, termed accessible cholesterol, functions as a second messenger that conveys the signal between PTCH1 and SMO. Beyond solving a central puzzle in Hh signaling, these studies are revealing new principles in membrane biology: how proteins respond to and remodel cholesterol accessibility in membranes and how the cholesterol composition of organelle membranes is used to regulate protein function.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Receptor Patched-1/metabolismo , Receptor Smoothened/metabolismo , Animales , Membrana Celular/química , Colesterol/química , Cilios/química , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células Epiteliales/química , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Proteínas Hedgehog/química , Proteínas Hedgehog/genética , Humanos , Receptor Patched-1/química , Receptor Patched-1/genética , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Receptor Smoothened/química , Receptor Smoothened/genética
10.
Nat Chem Biol ; 16(2): 206-213, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932720

RESUMEN

Genetic screens in cultured human cells represent a powerful unbiased strategy to identify cellular pathways that determine drug efficacy, providing critical information for clinical development. We used insertional mutagenesis-based screens in haploid cells to identify genes required for the sensitivity to lasonolide A (LasA), a macrolide derived from a marine sponge that kills certain types of cancer cells at low nanomolar concentrations. Our screens converged on a single gene, LDAH, encoding a member of the metabolite serine hydrolase family that is localized on the surface of lipid droplets. Mechanistic studies revealed that LasA accumulates in lipid droplets, where it is cleaved into a toxic metabolite by LDAH. We suggest that selective partitioning of hydrophobic drugs into the oil phase of lipid droplets can influence their activation and eventual toxicity to cells.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Gotas Lipídicas/metabolismo , Macrólidos/farmacocinética , Macrólidos/toxicidad , Proteínas/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Haploidia , Humanos , Inactivación Metabólica , Gotas Lipídicas/efectos de los fármacos , Macrólidos/metabolismo , Proteínas/genética
11.
Nature ; 535(7613): 517-522, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27437577

RESUMEN

Developmental signals of the Hedgehog (Hh) and Wnt families are transduced across the membrane by Frizzledclass G-protein-coupled receptors (GPCRs) composed of both a heptahelical transmembrane domain (TMD) and an extracellular cysteine-rich domain (CRD). How the large extracellular domains of GPCRs regulate signalling by the TMD is unknown. We present crystal structures of the Hh signal transducer and oncoprotein Smoothened, a GPCR that contains two distinct ligand-binding sites: one in its TMD and one in the CRD. The CRD is stacked a top the TMD, separated by an intervening wedge-like linker domain. Structure-guided mutations show that the interface between the CRD, linker domain and TMD stabilizes the inactive state of Smoothened. Unexpectedly, we find a cholesterol molecule bound to Smoothened in the CRD binding site. Mutations predicted to prevent cholesterol binding impair the ability of Smoothened to transmit native Hh signals. Binding of a clinically used antagonist, vismodegib, to the TMD induces a conformational change that is propagated to the CRD, resulting in loss of cholesterol from the CRD-linker domain-TMD interface. Our results clarify the structural mechanism by which the activity of a GPCR is controlled by ligand-regulated interactions between its extracellular and transmembrane domains.


Asunto(s)
Espacio Extracelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Anilidas/química , Anilidas/metabolismo , Anilidas/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión/genética , Colesterol/metabolismo , Colesterol/farmacología , Cristalografía por Rayos X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Ligandos , Modelos Moleculares , Unión Proteica/genética , Estabilidad Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Receptor Smoothened
12.
Nat Chem Biol ; 15(10): 975-982, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31548691

RESUMEN

Hedgehog (HH) ligands, classical morphogens that pattern embryonic tissues in all animals, are covalently coupled to two lipids-a palmitoyl group at the N terminus and a cholesteroyl group at the C terminus. While the palmitoyl group binds and inactivates Patched 1 (PTCH1), the main receptor for HH ligands, the function of the cholesterol modification has remained mysterious. Using structural and biochemical studies, along with reassessment of previous cryo-electron microscopy structures, we find that the C-terminal cholesterol attached to Sonic hedgehog (Shh) binds the first extracellular domain of PTCH1 and promotes its inactivation, thus triggering HH signaling. Molecular dynamics simulations show that this interaction leads to the closure of a tunnel through PTCH1 that serves as the putative conduit for sterol transport. Thus, Shh inactivates PTCH1 by grasping its extracellular domain with two lipidic pincers, the N-terminal palmitate and the C-terminal cholesterol, which are both inserted into the PTCH1 protein core.


Asunto(s)
Proteínas Hedgehog/metabolismo , Receptor Patched-1/metabolismo , Animales , Colesterol/química , Regulación de la Expresión Génica , Células HEK293 , Proteínas Hedgehog/química , Proteínas Hedgehog/genética , Humanos , Ratones , Modelos Moleculares , Células 3T3 NIH , Receptor Patched-1/química , Unión Proteica , Conformación Proteica , Anticuerpos de Dominio Único
13.
EMBO Rep ; 17(5): 739-52, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27113758

RESUMEN

The G-protein-coupled receptor kinase 2 (adrbk2/GRK2) has been implicated in vertebrate Hedgehog (Hh) signalling based on the effects of its transient knock-down in mammalian cells and zebrafish embryos. Here, we show that the response to Hh signalling is effectively abolished in the absence of Grk2 activity. Zebrafish embryos lacking all Grk2 activity are refractory to both Sonic hedgehog (Shh) and oncogenic Smoothened (Smo) activity, but remain responsive to inhibition of cAMP-dependent protein kinase (PKA) activity. Mutation of the kinase domain abrogates the rescuing activity of grk2 mRNA, suggesting that Grk2 acts in a kinase-dependent manner to regulate the response to Hh. Previous studies have suggested that Grk2 potentiates Smo activity by phosphorylating its C-terminal tail (CTT). In the zebrafish embryo, however, phosphomimetic Smo does not display constitutive activity, whereas phospho-null mutants retain activity, implying phosphorylation is neither sufficient nor necessary for Smo function. Since Grk2 rescuing activity requires the integrity of domains essential for its interaction with GPCRs, we speculate that Grk2 may regulate Hh pathway activity by downregulation of a GPCR.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Alelos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Análisis por Conglomerados , Activación Enzimática , Quinasa 2 del Receptor Acoplado a Proteína-G/química , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Técnicas de Inactivación de Genes , Células Germinativas/metabolismo , Humanos , Ratones , Mutación , Fenotipo , Fosforilación , Pez Cebra
15.
PLoS Genet ; 11(8): e1005473, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26291458

RESUMEN

The G protein-coupled receptor (GPCR) Smoothened (Smo) is the requisite signal transducer of the evolutionarily conserved Hedgehog (Hh) pathway. Although aspects of Smo signaling are conserved from Drosophila to vertebrates, significant differences have evolved. These include changes in its active sub-cellular localization, and the ability of vertebrate Smo to induce distinct G protein-dependent and independent signals in response to ligand. Whereas the canonical Smo signal to Gli transcriptional effectors occurs in a G protein-independent manner, its non-canonical signal employs Gαi. Whether vertebrate Smo can selectively bias its signal between these routes is not yet known. N-linked glycosylation is a post-translational modification that can influence GPCR trafficking, ligand responsiveness and signal output. Smo proteins in Drosophila and vertebrate systems harbor N-linked glycans, but their role in Smo signaling has not been established. Herein, we present a comprehensive analysis of Drosophila and murine Smo glycosylation that supports a functional divergence in the contribution of N-linked glycans to signaling. Of the seven predicted glycan acceptor sites in Drosophila Smo, one is essential. Loss of N-glycosylation at this site disrupted Smo trafficking and attenuated its signaling capability. In stark contrast, we found that all four predicted N-glycosylation sites on murine Smo were dispensable for proper trafficking, agonist binding and canonical signal induction. However, the under-glycosylated protein was compromised in its ability to induce a non-canonical signal through Gαi, providing for the first time evidence that Smo can bias its signal and that a post-translational modification can impact this process. As such, we postulate a profound shift in N-glycan function from affecting Smo ER exit in flies to influencing its signal output in mice.


Asunto(s)
Proteínas de Drosophila/metabolismo , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Drosophila melanogaster , Glicosilación , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Unión Proteica , Transporte de Proteínas , Transducción de Señal , Receptor Smoothened , Especificidad de la Especie
16.
Genes Dev ; 24(7): 670-82, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20360384

RESUMEN

The transcriptional program orchestrated by Hedgehog signaling depends on the Gli family of transcription factors. Gli proteins can be converted to either transcriptional activators or truncated transcriptional repressors. We show that the interaction between Gli3 and Suppressor of Fused (Sufu) regulates the formation of either repressor or activator forms of Gli3. In the absence of signaling, Sufu restrains Gli3 in the cytoplasm, promoting its processing into a repressor. Initiation of signaling triggers the dissociation of Sufu from Gli3. This event prevents formation of the repressor and instead allows Gli3 to enter the nucleus, where it is converted into a labile, differentially phosphorylated transcriptional activator. This key dissociation event depends on Kif3a, a kinesin motor required for the function of primary cilia. We propose that the Sufu-Gli3 interaction is a major control point in the Hedgehog pathway, a pathway that plays important roles in both development and cancer.


Asunto(s)
Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Cinesinas/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Proteína Gli3 con Dedos de Zinc
17.
Semin Cell Dev Biol ; 33: 63-72, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24845016

RESUMEN

The Hedgehog (Hh) pathway has become an important model to study the cell biology of primary cilia, and reciprocally, the study of ciliary processes provides an opportunity to solve longstanding mysteries in the mechanism of vertebrate Hh signal transduction. The cilium is emerging as an unique compartment for G-protein-coupled receptor (GPCR) signaling in many systems. Two members of the GPCR family, Smoothened and Gpr161, play important roles in the Hh pathway. We review the current understanding of how these proteins may function to regulate Hh signaling and also highlight some of the critical unanswered questions being tackled by the field. Uncovering GPCR-regulated mechanisms important in Hh signaling may provide therapeutic strategies against the Hh pathway that plays important roles in development, regeneration and cancer.


Asunto(s)
Cilios/fisiología , Proteínas Hedgehog/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Tipificación del Cuerpo , Humanos , Transducción de Señal
18.
J Biol Chem ; 289(16): 11095-11110, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24596093

RESUMEN

Oxysterols, oxidized metabolites of cholesterol, are endogenous small molecules that regulate lipid metabolism, immune function, and developmental signaling. Although the cell biology of cholesterol has been intensively studied, fundamental questions about oxysterols, such as their subcellular distribution and trafficking pathways, remain unanswered. We have therefore developed a useful method to image intracellular 20(S)-hydroxycholesterol with both high sensitivity and spatial resolution using click chemistry and fluorescence microscopy. The metabolic labeling of cells with an alkynyl derivative of 20(S)-hydroxycholesterol has allowed us to directly visualize this oxysterol by attaching an azide fluorophore through cyclo-addition. Unexpectedly, we found that this oxysterol selectively accumulates in the Golgi membrane using a pathway that is sensitive to ATP levels, temperature, and lysosome function. Although previous models have proposed nonvesicular pathways for the rapid equilibration of oxysterols between membranes, direct imaging of oxysterols suggests that a vesicular pathway is responsible for differential accumulation of oxysterols in organelle membranes. More broadly, clickable alkynyl sterols may represent useful tools for sterol cell biology, both to investigate the functions of these important lipids and to decipher the pathways that determine their cellular itineraries.


Asunto(s)
Química Clic , Colorantes Fluorescentes , Aparato de Golgi/metabolismo , Hidroxicolesteroles , Membranas Intracelulares/metabolismo , Animales , Transporte Biológico Activo/fisiología , Células CHO , Cricetinae , Cricetulus , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Hidroxicolesteroles/síntesis química , Hidroxicolesteroles/química , Hidroxicolesteroles/metabolismo , Ratones , Microscopía Fluorescente , Células 3T3 NIH
19.
Nat Chem Biol ; 9(7): 437-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23666116

RESUMEN

Primary cilia function as specialized compartments for signal transduction. The stereotyped structure and signaling function of cilia inextricably depend on the selective segregation of molecules in cilia. However, the fundamental principles governing the access of soluble proteins to primary cilia remain unresolved. We developed a methodology termed 'chemically inducible diffusion trap at cilia' to visualize the diffusion process of a series of fluorescent proteins ranging in size from 3.2 nm to 7.9 nm into primary cilia. We found that the interior of the cilium was accessible to proteins as large as 7.9 nm. The kinetics of ciliary accumulation of this panel of proteins was exponentially limited by their Stokes radii. Quantitative modeling suggests that the diffusion barrier operates as a molecular sieve at the base of cilia. Our study presents a set of powerful, generally applicable tools for the quantitative monitoring of ciliary protein diffusion under both physiological and pathological conditions.


Asunto(s)
Cilios/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Cromatografía en Gel , Citosol/metabolismo , Difusión , Dimerización , Colorantes Fluorescentes , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/metabolismo , Ratones , Microtúbulos/metabolismo , Modelos Químicos , Células 3T3 NIH , Espectrometría de Fluorescencia
20.
Nat Chem Biol ; 8(2): 211-20, 2012 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-22231273

RESUMEN

Oxysterols are a class of endogenous signaling molecules that can activate the Hedgehog pathway, which has critical roles in development, regeneration and cancer. However, it has been unclear how oxysterols influence Hedgehog signaling, including whether their effects are mediated through a protein target or indirectly through effects on membrane properties. To answer this question, we synthesized the enantiomer and an epimer of the most potent oxysterol, 20(S)-hydroxycholesterol. Using these molecules, we show that the effects of oxysterols on Hedgehog signaling are exquisitely stereoselective, consistent with the hypothesis that they function through a specific protein target. We present several lines of evidence that this protein target is the seven-pass transmembrane protein Smoothened, a major drug target in oncology. Our work suggests that these enigmatic sterols, which have multiple effects on cell physiology, may act as ligands for signaling receptors and provides a generally applicable framework for probing sterol signaling mechanisms.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroles/farmacología , Regulación Alostérica/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Humanos , Hidroxicolesteroles/síntesis química , Hidroxicolesteroles/química , Hidroxicolesteroles/farmacología , Ligandos , Proteínas Oncogénicas , Receptor Smoothened
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA