Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-37886503

RESUMEN

Homeostatic control of intracellular ionic strength is essential for protein, organelle and genome function, yet mechanisms that sense and enable adaptation to ionic stress remain poorly understood in animals. We find that the transcription factor NFAT5 directly senses solution ionic strength using a C-terminal intrinsically disordered region. Both in intact cells and in a purified system, NFAT5 forms dynamic, reversible biomolecular condensates in response to increasing ionic strength. This self-associative property, conserved from insects to mammals, allows NFAT5 to accumulate in the nucleus and activate genes that restore cellular ion content. Mutations that reduce condensation or those that promote aggregation both reduce NFAT5 activity, highlighting the importance of optimally tuned associative interactions. Remarkably, human NFAT5 alone is sufficient to reconstitute a mammalian transcriptional response to ionic or hypertonic stress in yeast. Thus NFAT5 is both the sensor and effector of a cell-autonomous ionic stress response pathway in animal cells.

2.
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
4.
EMBO Mol Med ; 12(11): e11739, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33200460

RESUMEN

Mutations in genes affecting primary cilia cause ciliopathies, a diverse group of disorders often affecting skeletal development. This includes Jeune syndrome or asphyxiating thoracic dystrophy (ATD), an autosomal recessive skeletal disorder. Unraveling the responsible molecular pathology helps illuminate mechanisms responsible for functional primary cilia. We identified two families with ATD caused by loss-of-function mutations in the gene encoding adrenergic receptor kinase 1 (ADRBK1 or GRK2). GRK2 cells from an affected individual homozygous for the p.R158* mutation resulted in loss of GRK2, and disrupted chondrocyte growth and differentiation in the cartilage growth plate. GRK2 null cells displayed normal cilia morphology, yet loss of GRK2 compromised cilia-based signaling of Hedgehog (Hh) pathway. Canonical Wnt signaling was also impaired, manifested as a failure to respond to Wnt ligand due to impaired phosphorylation of the Wnt co-receptor LRP6. We have identified GRK2 as an essential regulator of skeletogenesis and demonstrate how both Hh and Wnt signaling mechanistically contribute to skeletal ciliopathies.


Asunto(s)
Síndrome de Ellis-Van Creveld , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Proteínas Hedgehog , Proteínas Hedgehog/genética , Humanos , Mutación , Vía de Señalización Wnt
5.
Dev Cell ; 55(4): 432-449.e12, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32966817

RESUMEN

The etiology of congenital heart defects (CHDs), which are among the most common human birth defects, is poorly understood because of its complex genetic architecture. Here, we show that two genes implicated in CHDs, Megf8 and Mgrn1, interact genetically and biochemically to regulate the strength of Hedgehog signaling in target cells. MEGF8, a transmembrane protein, and MGRN1, a RING superfamily E3 ligase, assemble to form a receptor-like ubiquitin ligase complex that catalyzes the ubiquitination and degradation of the Hedgehog pathway transducer Smoothened. Homozygous Megf8 and Mgrn1 mutations increased Smoothened abundance and elevated sensitivity to Hedgehog ligands. While mice heterozygous for loss-of-function Megf8 or Mgrn1 mutations were normal, double heterozygous embryos exhibited an incompletely penetrant syndrome of CHDs with heterotaxy. Thus, genetic interactions can arise from biochemical mechanisms that calibrate morphogen signaling strength, a conclusion broadly relevant for the many human diseases in which oligogenic inheritance is emerging as a mechanism for heritability.


Asunto(s)
Corazón/embriología , Proteínas Hedgehog/metabolismo , Transducción de Señal , Ubiquitinación , Alelos , Animales , Embrión de Mamíferos/metabolismo , Epistasis Genética , Dosificación de Gen , Proteínas de la Membrana/metabolismo , Ratones , Mutación/genética , Células 3T3 NIH , Fenotipo , Unión Proteica , Receptor Smoothened/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Elife ; 92020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32432544

RESUMEN

R-spondins (RSPOs) amplify WNT signaling during development and regenerative responses. We previously demonstrated that RSPOs 2 and 3 potentiate WNT/ß-catenin signaling in cells lacking leucine-rich repeat-containing G-protein coupled receptors (LGRs) 4, 5 and 6 (Lebensohn and Rohatgi, 2018). We now show that heparan sulfate proteoglycans (HSPGs) act as alternative co-receptors for RSPO3 using a combination of ligand mutagenesis and ligand engineering. Mutations in RSPO3 residues predicted to contact HSPGs impair its signaling capacity. Conversely, the HSPG-binding domains of RSPO3 can be entirely replaced with an antibody that recognizes heparan sulfate (HS) chains attached to multiple HSPGs without diminishing WNT-potentiating activity in cultured cells and intestinal organoids. A genome-wide screen for mediators of RSPO3 signaling in cells lacking LGRs 4, 5 and 6 failed to reveal other receptors. We conclude that HSPGs are RSPO co-receptors that potentiate WNT signaling in the presence and absence of LGRs.


Asunto(s)
Proteoglicanos de Heparán Sulfato/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vía de Señalización Wnt , Células Cultivadas , Biología Evolutiva , Proteoglicanos de Heparán Sulfato/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ligandos , Organoides , Receptores Acoplados a Proteínas G/genética , Trombospondinas
7.
Elife ; 82019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31657721

RESUMEN

Previously we proposed that transmission of the hedgehog signal across the plasma membrane by Smoothened is triggered by its interaction with cholesterol (Luchetti et al., 2016). But how is cholesterol, an abundant lipid, regulated tightly enough to control a signaling system that can cause birth defects and cancer? Using toxin-based sensors that distinguish between distinct pools of cholesterol, we find that Smoothened activation and Hedgehog signaling are driven by a biochemically-defined, small fraction of membrane cholesterol, termed accessible cholesterol. Increasing cholesterol accessibility by depletion of sphingomyelin, which sequesters cholesterol in complexes, amplifies Hedgehog signaling. Hedgehog ligands increase cholesterol accessibility in the membrane of the primary cilium by inactivating the transporter-like protein Patched 1. Trapping this accessible cholesterol blocks Hedgehog signal transmission across the membrane. Our work shows that the organization of cholesterol in the ciliary membrane can be modified by extracellular ligands to control the activity of cilia-localized signaling proteins.


Asunto(s)
Colesterol/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos
9.
PLoS One ; 14(1): e0198463, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30695034

RESUMEN

The systematic identification of regulatory elements that control gene expression remains a challenge. Genetic screens that use untargeted mutagenesis have the potential to identify protein-coding genes, non-coding RNAs and regulatory elements, but their analysis has mainly focused on identifying the former two. To identify regulatory elements, we conducted a new bioinformatics analysis of insertional mutagenesis screens interrogating WNT signaling in haploid human cells. We searched for specific patterns of retroviral gene trap integrations (used as mutagens in haploid screens) in short genomic intervals overlapping with introns and regions upstream of genes. We uncovered atypical patterns of gene trap insertions that were not predicted to disrupt coding sequences, but caused changes in the expression of two key regulators of WNT signaling, suggesting the presence of cis-regulatory elements. Our methodology extends the scope of haploid genetic screens by enabling the identification of regulatory elements that control gene expression.


Asunto(s)
Intrones , Modelos Genéticos , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ADN , Animales , Biología Computacional , Genómica , Haploidia , Humanos , Mutagénesis Insercional , Retroviridae/genética
10.
Nat Commun ; 9(1): 1732, 2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712905

RESUMEN

Tubulin post-translational modifications (PTMs) occur spatiotemporally throughout cells and are suggested to be involved in a wide range of cellular activities. However, the complexity and dynamic distribution of tubulin PTMs within cells have hindered the understanding of their physiological roles in specific subcellular compartments. Here, we develop a method to rapidly deplete tubulin glutamylation inside the primary cilia, a microtubule-based sensory organelle protruding on the cell surface, by targeting an engineered deglutamylase to the cilia in minutes. This rapid deglutamylation quickly leads to altered ciliary functions such as kinesin-2-mediated anterograde intraflagellar transport and Hedgehog signaling, along with no apparent crosstalk to other PTMs such as acetylation and detyrosination. Our study offers a feasible approach to spatiotemporally manipulate tubulin PTMs in living cells. Future expansion of the repertoire of actuators that regulate PTMs may facilitate a comprehensive understanding of how diverse tubulin PTMs encode ciliary as well as cellular functions.


Asunto(s)
Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo , Acetilación , Animales , Cilios/ultraestructura , Genes Reporteros , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Hedgehog/genética , Cinesinas , Ratones , Proteínas Asociadas a Microtúbulos/genética , Células 3T3 NIH , Imagen Óptica , Transducción de Señal , Tubulina (Proteína)/genética , Tirosina/metabolismo
11.
Sci Signal ; 11(516)2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29438014

RESUMEN

The morphogen Sonic Hedgehog (SHH) patterns tissues during development by directing cell fates in a concentration-dependent manner. The SHH signal is transmitted across the membrane of target cells by the heptahelical transmembrane protein Smoothened (SMO), which activates the GLI family of transcription factors through a mechanism that is undefined in vertebrates. Using CRISPR-edited null alleles and small-molecule inhibitors, we systematically analyzed the epistatic interactions between SMO and three proteins implicated in SMO signaling: the heterotrimeric G protein subunit GαS, the G protein-coupled receptor kinase 2 (GRK2), and the GαS-coupled receptor GPR161. Our experiments uncovered a signaling mechanism that modifies the sensitivity of target cells to SHH and consequently changes the shape of the SHH dose-response curve. In both fibroblasts and spinal neural progenitors, the loss of GPR161, previously implicated as an inhibitor of basal SHH signaling, increased the sensitivity of target cells across the entire spectrum of SHH concentrations. Even in cells lacking GPR161, GRK2 was required for SHH signaling, and Gαs, which promotes the activation of protein Kinase A (PKA), antagonized SHH signaling. We propose that the sensitivity of target cells to Hedgehog morphogens, and the consequent effects on gene expression and differentiation outcomes, can be controlled by signals from G protein-coupled receptors that converge on Gαs and PKA.


Asunto(s)
Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Animales , Células Cultivadas , Cromograninas/genética , Cromograninas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epistasis Genética , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Ratones , Células 3T3 NIH , Receptores Acoplados a Proteínas G/genética , Receptor Smoothened/genética
13.
Dev Cell ; 44(1): 113-129.e8, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29290584

RESUMEN

To uncover regulatory mechanisms in Hedgehog (Hh) signaling, we conducted genome-wide screens to identify positive and negative pathway components and validated top hits using multiple signaling and differentiation assays in two different cell types. Most positive regulators identified in our screens, including Rab34, Pdcl, and Tubd1, were involved in ciliary functions, confirming the central role for primary cilia in Hh signaling. Negative regulators identified included Megf8, Mgrn1, and an unannotated gene encoding a tetraspan protein we named Atthog. The function of these negative regulators converged on Smoothened (SMO), an oncoprotein that transduces the Hh signal across the membrane. In the absence of Atthog, SMO was stabilized at the cell surface and concentrated in the ciliary membrane, boosting cell sensitivity to the ligand Sonic Hedgehog (SHH) and consequently altering SHH-guided neural cell-fate decisions. Thus, we uncovered genes that modify the interpretation of morphogen signals by regulating protein-trafficking events in target cells.


Asunto(s)
Cilios/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteínas Hedgehog/farmacología , Neuronas/fisiología , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Cilios/efectos de los fármacos , Fluorescencia , Proteínas de la Membrana/metabolismo , Ratones , Células 3T3 NIH , Neuronas/citología , Neuronas/efectos de los fármacos , Receptor Smoothened/genética
14.
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
15.
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
16.
Nature ; 521(7550): 99-104, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25924063

RESUMEN

Whereas cancers grow within host tissues and evade host immunity through immune-editing and immunosuppression, tumours are rarely transmissible between individuals. Much like transplanted allogeneic organs, allogeneic tumours are reliably rejected by host T cells, even when the tumour and host share the same major histocompatibility complex alleles, the most potent determinants of transplant rejection. How such tumour-eradicating immunity is initiated remains unknown, although elucidating this process could provide the basis for inducing similar responses against naturally arising tumours. Here we find that allogeneic tumour rejection is initiated in mice by naturally occurring tumour-binding IgG antibodies, which enable dendritic cells (DCs) to internalize tumour antigens and subsequently activate tumour-reactive T cells. We exploited this mechanism to treat autologous and autochthonous tumours successfully. Either systemic administration of DCs loaded with allogeneic-IgG-coated tumour cells or intratumoral injection of allogeneic IgG in combination with DC stimuli induced potent T-cell-mediated antitumour immune responses, resulting in tumour eradication in mouse models of melanoma, pancreas, lung and breast cancer. Moreover, this strategy led to eradication of distant tumours and metastases, as well as the injected primary tumours. To assess the clinical relevance of these findings, we studied antibodies and cells from patients with lung cancer. T cells from these patients responded vigorously to autologous tumour antigens after culture with allogeneic-IgG-loaded DCs, recapitulating our findings in mice. These results reveal that tumour-binding allogeneic IgG can induce powerful antitumour immunity that can be exploited for cancer immunotherapy.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Inmunoglobulina G/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Antineoplásicos/administración & dosificación , Antígenos CD40/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/administración & dosificación , Isoanticuerpos/administración & dosificación , Isoanticuerpos/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias/inmunología , Neoplasias/patología , Receptores de IgG/inmunología , Factor de Necrosis Tumoral alfa/inmunología
18.
Dev Cell ; 28(5): 483-96, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24582806

RESUMEN

The Hedgehog (Hh) pathway depends on primary cilia in vertebrates, but the signaling machinery within cilia remains incompletely defined. We report the identification of a complex between two ciliary proteins, EFCAB7 and IQCE, which positively regulates the Hh pathway. The EFCAB7-IQCE module anchors the EVC-EVC2 complex in a signaling microdomain at the base of cilia. EVC and EVC2 genes are mutated in Ellis van Creveld and Weyers syndromes, characterized by impaired Hh signaling in skeletal, cardiac, and orofacial tissues. EFCAB7 binds to a C-terminal disordered region in EVC2 that is deleted in Weyers patients. EFCAB7 depletion mimics the Weyers cellular phenotype-the mislocalization of EVC-EVC2 within cilia and impaired activation of the transcription factor GLI2. Evolutionary analysis suggests that emergence of these complexes might have been important for adaptation of an ancient organelle, the cilium, for an animal-specific signaling network.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Cilios/fisiología , Regulación de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de la Membrana/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proteínas de Unión al Calcio/genética , Cromatografía de Afinidad , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Mutación/genética , Células 3T3 NIH , Fenotipo , Filogenia , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Transducción de Señal , Receptor Smoothened , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteína Gli2 con Dedos de Zinc
19.
J Biol Chem ; 287(50): 42129-37, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23091056

RESUMEN

Rab GTPases are master regulators of membrane trafficking events and template the directionality of protein transport through the secretory and endocytic pathways. Certain Rabs recruit the guanine nucleotide exchange factor (GEF) that activates a subsequent acting Rab protein in a given pathway; this process has been termed a Rab cascade. We show here that the medial Golgi-localized Rab33B GTPase has the potential to link functionally to the late Golgi, Rab6 GTPase, by its capacity for association with Ric1 and Rgp1 proteins. In yeast, Ric1p and Rgp1p form a complex that catalyzes guanine nucleotide exchange by Ypt6p, the Rab6 homolog. Human Ric1 and Rgp1 both bind Rab6A with preference for the GDP-bound conformation, characteristic of a GEF. Nevertheless, both Ric1 and Rgp1 proteins are needed to catalyze nucleotide exchange on Rab6A protein. Ric1 and Rgp1 form a complex, but unlike their yeast counterparts, most of the subunits are not associated, and most of the proteins are cytosolic. Loss of Ric1 or Rgp1 leads to destabilization of Rab6, concomitant with a block in Rab6-dependent retrograde transport of mannose 6-phosphate receptors to the Golgi. The C terminus of Ric1 protein contains a distinct binding site for Rab33B-GTP, supporting the existence of a Rab cascade between the medial and trans Golgi. This study thus identifies a GEF for Rab6A in human cells.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Estabilidad de Enzimas/fisiología , Factores de Intercambio de Guanina Nucleótido/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Complejos Multiproteicos/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas de Unión al GTP rab/genética , Red trans-Golgi/genética , Red trans-Golgi/metabolismo
20.
J Biol Chem ; 287(27): 22740-8, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22637480

RESUMEN

Rab GTPases regulate vesicle budding, motility, docking, and fusion. In cells, their cycling between active, GTP-bound states and inactive, GDP-bound states is regulated by the action of opposing enzymes called guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). The substrates for most RabGAPs are unknown, and the potential for cross-talk between different membrane trafficking pathways remains uncharted territory. Rab9A and its effectors regulate recycling of mannose 6-phosphate receptors from late endosomes to the trans Golgi network. We show here that RUTBC2 is a TBC domain-containing protein that binds to Rab9A specifically both in vitro and in cultured cells but is not a GAP for Rab9A. Biochemical screening of Rab protein substrates for RUTBC2 revealed highest GAP activity toward Rab34 and Rab36. In cells, membrane-associated RUTBC2 co-localizes with Rab36, and expression of wild type RUTBC2, but not the catalytically inactive, RUTBC2 R829A mutant, decreases the amount of membrane-associated Rab36 protein. These data show that RUTBC2 can act as a Rab36 GAP in cells and suggest that RUTBC2 links Rab9A function to Rab36 function in the endosomal system.


Asunto(s)
Endosomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transporte de Proteínas/fisiología , Proteínas de Unión al GTP rab/metabolismo , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Hidrólisis , Péptidos y Proteínas de Señalización Intracelular/genética , Datos de Secuencia Molecular , Neuroblastoma , Neuronas/citología , Neuronas/metabolismo , Técnicas del Sistema de Dos Híbridos , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...