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.
ACS Chem Neurosci ; 15(1): 98-107, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38096481

RESUMEN

Prion diseases are fatal neurodegenerative disorders characterized by the conversion of the cellular prion protein (PrPC) into a misfolded prion form, which is believed to disrupt the cellular membranes. However, the exact mechanisms underlying prion toxicity, including the formation of membrane pores, are not fully understood. The prion protein consists of two domains: a globular domain (GD) and a flexible N-terminus (FT) domain. Although a proximal polybasic amino acid (FT(23-31) sequence of FT is a prerequisite for cellular membrane permeabilization, other functional domain regions may modulate its effects. Through single-channel electrical recordings and cryo-electron microscopy (cryo-EM), we discovered that the FT(23-50) fragment forms pore-shaped oligomers and plays a dominant role in membrane permeabilization within the full-length mouse prion protein (mPrP(23-230)). In contrast, the FT(51-110) domain or the C-terminal domain downregulate the channel activity of FT(23-50) and mPrP(23-230). The addition of prion mimetic antibody, POM1 significantly amplifies mPrP(23-230) membrane permeabilization, whereas POM1_Y104A, a mutant that binds to PrP but cannot elicit toxicity, has a negligible effect on membrane permeabilization. Additionally, the anti-N-terminal antibody POM2 or Cu2+ binds to the FT domain, subsequently enhancing the FT(23-110) channel activity. Importantly, our setup provides a novel approach without an external fused protein to examine the channel activity of truncated PrP in the lipid membranes. We therefore propose that the primary N-terminal residues are essential for membrane permeabilization, while other functional segments of PrP play a vital role in modulating the pathological effects of PrP-mediated neurotoxicity.


Asunto(s)
Proteínas PrPC , Enfermedades por Prión , Priones , Ratones , Animales , Priones/metabolismo , Proteínas Priónicas/genética , Microscopía por Crioelectrón , Membrana Celular/metabolismo , Anticuerpos , Proteínas PrPC/química
2.
Cell Rep ; 41(8): 111689, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36417879

RESUMEN

Calreticulin (CALR) is an endoplasmic reticulum (ER)-retained chaperone that assists glycoproteins in obtaining their structure. CALR mutations occur in patients with myeloproliferative neoplasms (MPNs), and the ER retention of CALR mutants (CALR MUT) is reduced due to a lacking KDEL sequence. Here, we investigate the impact of CALR mutations on protein structure and protein levels in MPNs by subjecting primary patient samples and CALR-mutated cell lines to limited proteolysis-coupled mass spectrometry (LiP-MS). Especially glycoproteins are differentially expressed and undergo profound structural alterations in granulocytes and cell lines with homozygous, but not with heterozygous, CALR mutations. Furthermore, homozygous CALR mutations and loss of CALR equally perturb glycoprotein integrity, suggesting that loss-of-function attributes of mutated CALR chaperones (CALR MUT) lead to glycoprotein maturation defects. Finally, by investigating the misfolding of the CALR glycoprotein client myeloperoxidase (MPO), we provide molecular proof of protein misfolding in the presence of homozygous CALR mutations.


Asunto(s)
Calreticulina , Trastornos Mieloproliferativos , Humanos , Calreticulina/genética , Calreticulina/química , Calreticulina/metabolismo , Mutación/genética , Homocigoto , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteoma/metabolismo
3.
EMBO J ; 41(23): e112338, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36254605

RESUMEN

A defining characteristic of mammalian prions is their capacity for self-sustained propagation. Theoretical considerations and experimental evidence suggest that prion propagation is modulated by cell-autonomous and non-autonomous modifiers. Using a novel quantitative phospholipase protection assay (QUIPPER) for high-throughput prion measurements, we performed an arrayed genome-wide RNA interference (RNAi) screen aimed at detecting cellular host-factors that can modify prion propagation. We exposed prion-infected cells in high-density microplates to 35,364 ternary pools of 52,746 siRNAs targeting 17,582 genes representing the majority of the mouse protein-coding transcriptome. We identified 1,191 modulators of prion propagation. While 1,151 modified the expression of both the pathological prion protein, PrPSc , and its cellular counterpart, PrPC , 40 genes selectively affected PrPSc . Of the latter 40 genes, 20 augmented prion production when suppressed. A prominent limiter of prion propagation was the heterogeneous nuclear ribonucleoprotein Hnrnpk. Psammaplysene A (PSA), which binds Hnrnpk, reduced prion levels in cultured cells and protected them from cytotoxicity. PSA also reduced prion levels in infected cerebellar organotypic slices and alleviated locomotor deficits in prion-infected Drosophila melanogaster expressing ovine PrPC . Hence, genome-wide QUIPPER-based perturbations can discover actionable cellular pathways involved in prion propagation. Further, the unexpected identification of a prion-controlling ribonucleoprotein suggests a role for RNA in the generation of infectious prions.


Asunto(s)
Enfermedades por Prión , Priones , Ratones , Animales , Ovinos/genética , Priones/genética , Priones/metabolismo , Drosophila melanogaster/genética , Ribonucleoproteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/patología , Mamíferos/genética
4.
Nat Struct Mol Biol ; 29(8): 831-840, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948768

RESUMEN

Prion infections cause conformational changes of the cellular prion protein (PrPC) and lead to progressive neurological impairment. Here we show that toxic, prion-mimetic ligands induce an intramolecular R208-H140 hydrogen bond ('H-latch'), altering the flexibility of the α2-α3 and ß2-α2 loops of PrPC. Expression of a PrP2Cys mutant mimicking the H-latch was constitutively toxic, whereas a PrPR207A mutant unable to form the H-latch conferred resistance to prion infection. High-affinity ligands that prevented H-latch induction repressed prion-related neurodegeneration in organotypic cerebellar cultures. We then selected phage-displayed ligands binding wild-type PrPC, but not PrP2Cys. These binders depopulated H-latched conformers and conferred protection against prion toxicity. Finally, brain-specific expression of an antibody rationally designed to prevent H-latch formation prolonged the life of prion-infected mice despite unhampered prion propagation, confirming that the H-latch is an important reporter of prion neurotoxicity.


Asunto(s)
Proteínas PrPC , Priones , Animales , Anticuerpos/metabolismo , Cerebelo/metabolismo , Ligandos , Ratones , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas Priónicas/química , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Priones/metabolismo , Priones/toxicidad
5.
Brain Pathol ; 32(5): e13056, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35178783

RESUMEN

Although prion infections cause cognitive impairment and neuronal death, transcriptional and translational profiling shows progressive derangement within glia but surprisingly little changes within neurons. Here we expressed PrPC selectively in neurons and astrocytes of mice. After prion infection, both astrocyte and neuron-restricted PrPC expression led to copious brain accumulation of PrPSc . As expected, neuron-restricted expression was associated with typical prion disease. However, mice with astrocyte-restricted PrPC expression experienced a normal life span, did not develop clinical disease, and did not show astro- or microgliosis. Besides confirming that PrPSc is innocuous to PrPC -deficient neurons, these results show that astrocyte-born PrPSc does not activate the extreme neuroinflammation that accompanies the onset of prion disease and precedes any molecular changes of neurons. This points to a nonautonomous mechanism by which prion-infected neurons instruct astrocytes and microglia to acquire a specific cellular state that, in turn, drives neural dysfunction.


Asunto(s)
Enfermedades por Prión , Priones , Animales , Astrocitos/metabolismo , Ratones , Neuroglía/metabolismo , Neuronas/metabolismo , Enfermedades por Prión/metabolismo , Priones/metabolismo
6.
EMBO Mol Med ; 13(9): e14714, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34291577

RESUMEN

Brain-matter vacuolation is a defining trait of all prion diseases, yet its cause is unknown. Here, we report that prion infection and prion-mimetic antibodies deplete the phosphoinositide kinase PIKfyve-which controls endolysosomal maturation-from mouse brains, cultured cells, organotypic brain slices, and brains of Creutzfeldt-Jakob disease victims. We found that PIKfyve is acylated by the acyltransferases zDHHC9 and zDHHC21, whose juxtavesicular topology is disturbed by prion infection, resulting in PIKfyve deacylation and rapid degradation, as well as endolysosomal hypertrophy and activation of TFEB-dependent lysosomal enzymes. A protracted unfolded protein response (UPR), typical of prion diseases, also induced PIKfyve deacylation and degradation. Conversely, UPR antagonists restored PIKfyve levels in prion-infected cells. Overexpression of zDHHC9 and zDHHC21, administration of the antiprion polythiophene LIN5044, or supplementation with the PIKfyve reaction product PI(3,5)P2 suppressed prion-induced vacuolation and restored lysosomal homeostasis. Thus, PIKfyve emerges as a central mediator of vacuolation and neurotoxicity in prion diseases.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Enfermedades por Prión , Aciltransferasas , Animales , Encéfalo/metabolismo , Homeostasis , Lisosomas/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo
7.
PLoS One ; 15(11): e0242137, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33180885

RESUMEN

The adhesion G-protein coupled receptor Adgrg6 (formerly Gpr126) is instrumental in the development, maintenance and repair of peripheral nervous system myelin. The prion protein (PrP) is a potent activator of Adgrg6 and could be used as a potential therapeutic agent in treating peripheral demyelinating and dysmyelinating diseases. We designed a dimeric Fc-fusion protein comprising the myelinotrophic domain of PrP (FT2Fc), which activated Adgrg6 in vitro and exhibited favorable pharmacokinetic properties for in vivo treatment of peripheral neuropathies. While chronic FT2Fc treatment elicited specific transcriptomic changes in the sciatic nerves of PrP knockout mice, no amelioration of the early molecular signs demyelination was detected. Instead, RNA sequencing of sciatic nerves revealed downregulation of cytoskeletal and sarcomere genes, akin to the gene expression changes seen in myopathic skeletal muscle of PrP overexpressing mice. These results call for caution when devising myelinotrophic therapies based on PrP-derived Adgrg6 ligands. While our treatment approach was not successful, Adgrg6 remains an attractive therapeutic target to be addressed in other disease models or by using different biologically active Adgrg6 ligands.


Asunto(s)
Enfermedades Desmielinizantes/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Proteínas Priónicas/química , Receptores Acoplados a Proteínas G/agonistas , Animales , Línea Celular , Enfermedades Desmielinizantes/genética , Femenino , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Proteínas Priónicas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Nervio Ciático/metabolismo , Transcriptoma
8.
J Exp Med ; 215(3): 941-961, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29367382

RESUMEN

Myelin is a multilamellar sheath generated by specialized glia called Schwann cells (SCs) in the peripheral nervous system (PNS), which serves to protect and insulate axons for rapid neuronal signaling. In zebrafish and rodent models, we identify GPR56/ADGRG1 as a conserved regulator of PNS development and health. We demonstrate that, during SC development, GPR56-dependent RhoA signaling promotes timely radial sorting of axons. In the mature PNS, GPR56 is localized to distinct SC cytoplasmic domains, is required to establish proper myelin thickness, and facilitates organization of the myelin sheath. Furthermore, we define plectin-a scaffolding protein previously linked to SC domain organization, myelin maintenance, and a series of disorders termed "plectinopathies"-as a novel interacting partner of GPR56. Finally, we show that Gpr56 mutants develop progressive neuropathy-like symptoms, suggesting an underlying mechanism for peripheral defects in some human patients with GPR56 mutations. In sum, we define Gpr56 as a new regulator in the development and maintenance of peripheral myelin.


Asunto(s)
Vaina de Mielina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Pez Cebra/fisiología , Animales , Citoesqueleto/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Mutación/genética , Vaina de Mielina/ultraestructura , Plectina/metabolismo , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Células de Schwann/metabolismo , Nervio Ciático/metabolismo , Transducción de Señal , Pez Cebra , Proteínas de Pez Cebra/genética , Proteína de Unión al GTP rhoA/metabolismo
9.
Annu Rev Pharmacol Toxicol ; 58: 331-351, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28961066

RESUMEN

Three decades after the discovery of prions as the cause of Creutzfeldt-Jakob disease and other transmissible spongiform encephalopathies, we are still nowhere close to finding an effective therapy. Numerous pharmacological interventions have attempted to target various stages of disease progression, yet none has significantly ameliorated the course of disease. We still lack a mechanistic understanding of how the prions damage the brain, and this situation results in a dearth of validated pharmacological targets. In this review, we discuss the attempts to interfere with the replication of prions and to enhance their clearance. We also trace some of the possibilities to identify novel targets that may arise with increasing insights into prion biology.


Asunto(s)
Enfermedades por Prión/tratamiento farmacológico , Priones/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Humanos
10.
PLoS Pathog ; 13(11): e1006733, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29176838

RESUMEN

Prion infections cause inexorable, progressive neurological dysfunction and neurodegeneration. Expression of the cellular prion protein PrPC is required for toxicity, suggesting the existence of deleterious PrPC-dependent signaling cascades. Because group-I metabotropic glutamate receptors (mGluR1 and mGluR5) can form complexes with the cellular prion protein (PrPC), we investigated the impact of mGluR1 and mGluR5 inhibition on prion toxicity ex vivo and in vivo. We found that pharmacological inhibition of mGluR1 and mGluR5 antagonized dose-dependently the neurotoxicity triggered by prion infection and by prion-mimetic anti-PrPC antibodies in organotypic brain slices. Prion-mimetic antibodies increased mGluR5 clustering around dendritic spines, mimicking the toxicity of Aß oligomers. Oral treatment with the mGluR5 inhibitor, MPEP, delayed the onset of motor deficits and moderately prolonged survival of prion-infected mice. Although group-I mGluR inhibition was not curative, these results suggest that it may alleviate the neurological dysfunctions induced by prion diseases.


Asunto(s)
Proteínas PrPC/toxicidad , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/metabolismo , Piridinas/administración & dosificación , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Anticuerpos/administración & dosificación , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/genética , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Receptor del Glutamato Metabotropico 5/genética , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo
11.
Nature ; 536(7617): 464-8, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27501152

RESUMEN

Ablation of the cellular prion protein PrP(C) leads to a chronic demyelinating polyneuropathy affecting Schwann cells. Neuron-restricted expression of PrP(C) prevents the disease, suggesting that PrP(C) acts in trans through an unidentified Schwann cell receptor. Here we show that the cAMP concentration in sciatic nerves from PrP(C)-deficient mice is reduced, suggesting that PrP(C) acts via a G protein-coupled receptor (GPCR). The amino-terminal flexible tail (residues 23-120) of PrP(C) triggered a concentration-dependent increase in cAMP in primary Schwann cells, in the Schwann cell line SW10, and in HEK293T cells overexpressing the GPCR Adgrg6 (also known as Gpr126). By contrast, naive HEK293T cells and HEK293T cells expressing several other GPCRs did not react to the flexible tail, and ablation of Gpr126 from SW10 cells abolished the flexible tail-induced cAMP response. The flexible tail contains a polycationic cluster (KKRPKPG) similar to the GPRGKPG motif of the Gpr126 agonist type-IV collagen. A KKRPKPG-containing PrPC-derived peptide (FT(23-50)) sufficed to induce a Gpr126-dependent cAMP response in cells and mice, and improved myelination in hypomorphic gpr126 mutant zebrafish (Danio rerio). Substitution of the cationic residues with alanines abolished the biological activity of both FT(23-50) and the equivalent type-IV collagen peptide. We conclude that PrP(C) promotes myelin homeostasis through flexible tail-mediated Gpr126 agonism. As well as clarifying the physiological role of PrP(C), these observations are relevant to the pathogenesis of demyelinating polyneuropathies--common debilitating diseases for which there are limited therapeutic options.


Asunto(s)
Priones/metabolismo , Priones/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Colágeno Tipo IV/química , Colágeno Tipo IV/farmacología , AMP Cíclico/metabolismo , Enfermedades Desmielinizantes/metabolismo , Femenino , Células HEK293 , Homeostasis/efectos de los fármacos , Humanos , Ligandos , Ratones , Datos de Secuencia Molecular , Vaina de Mielina/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Docilidad , Proteínas Priónicas , Priones/química , Priones/genética , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
12.
Blood ; 127(25): 3253-9, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27013444

RESUMEN

The pathogenesis of acquired myeloperoxidase (MPO) deficiency, a rare phenomenon observed in patients with Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), is unknown. MPO is a glycoprotein (GP) chaperoned by calreticulin (CALR) in the endoplasmic reticulum. Mutations in CALR are frequently found in patients with myelofibrosis (MF) and essential thrombocythemia (ET) with nonmutated Janus kinase 2 (JAK2). We hypothesized that acquired MPO deficiency in MPN could be associated with the presence of CALR mutations. A cohort of 317 patients with MPN (142 polycythemia vera [PV], 94 ET, and 81 MF) was screened for MPO deficiency. MPO deficiency was observed in 6/81 MF patients (7.4%), but not in PV or ET patients. Susceptibility to infections had been documented in 2/6 (33%) MPO-deficient patients. Five out of 6 patients with MPO deficiency carried a homozygous CALR mutation and were also deficient in eosinophilic peroxidase (EPX). In contrast, 1 patient with MF, a JAK2-V617F mutation, and MPO deficiency, carried 2 previously reported MPO mutations and showed normal EPX activity. Patients with homozygous CALR mutations had reduced MPO protein, but normal MPO messenger RNA (mRNA) levels supporting a posttranscriptional defect in MPO production. Finally, we demonstrate in vitro that in the absence of CALR, immature MPO protein precursors are degraded in the proteasome. Therefore, 4 decades after the first description of acquired MPO deficiency in MPN, we provide the molecular correlate associated with this phenomenon and evidence that CALR mutations can affect the biosynthesis of GPs.


Asunto(s)
Calreticulina/genética , Errores Innatos del Metabolismo/genética , Mutación , Mielofibrosis Primaria/genética , Animales , Células Cultivadas , Estudios de Cohortes , Homocigoto , Humanos , Errores Innatos del Metabolismo/patología , Ratones , Ratones Noqueados , Peroxidasa/genética , Peroxidasa/metabolismo , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
13.
Trends Cell Biol ; 26(1): 40-51, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26455408

RESUMEN

The coalescence of proteins into highly ordered aggregates is a hallmark of protein misfolding disorders (PMDs), which, when affecting the central nervous system, lead to progressive neurodegeneration. Although the chemical identity and the topology of each culprit protein are unique, the principles governing aggregation and propagation are strikingly stereotypical. It is now clear that such protein aggregates can spread from cell to cell and eventually affect entire organ systems - similarly to prion diseases. However, because most aggregates are not found to transmit between individuals, they are not infectious sensu strictiori. Therefore, they are not identical to prions and we prefer to define them as 'prionoids'. Here we review recent advances in understanding the toxicity of protein aggregation affecting the brain.


Asunto(s)
Proteínas Priónicas/fisiología , Péptidos beta-Amiloides/fisiología , Animales , Humanos , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Pliegue de Proteína , Multimerización de Proteína , Transporte de Proteínas , Deficiencias en la Proteostasis/metabolismo , Deficiencias en la Proteostasis/patología , Proteínas tau/fisiología
14.
PLoS One ; 10(2): e0117412, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25658480

RESUMEN

The cellular prion protein (PrPC) consists of a flexible N-terminal tail (FT, aa 23-128) hinged to a membrane-anchored globular domain (GD, aa 129-231). Ligation of the GD with antibodies induces rapid neurodegeneration, which is prevented by deletion or functional inactivation of the FT. Therefore, the FT is an allosteric effector of neurotoxicity. To explore its mechanism of action, we generated transgenic mice expressing the FT fused to a GPI anchor, but lacking the GD (PrPΔ141-225, or "FTgpi"). Here we report that FTgpi mice develop a progressive, inexorably lethal neurodegeneration morphologically and biochemically similar to that triggered by anti-GD antibodies. FTgpi was mostly retained in the endoplasmic reticulum, where it triggered a conspicuous unfolded protein response specifically activating the PERK pathway leading to phosphorylation of eIF2α and upregulation of CHOP ultimately leading to neurodegeration similar to what was observed in prion infection.


Asunto(s)
Cerebelo/patología , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Respuesta de Proteína Desplegada , Animales , Cerebelo/metabolismo , Estrés del Retículo Endoplásmico , Ratones , Ratones Transgénicos , Proteínas PrPC/análisis , Priones
15.
Mol Cell ; 51(3): 386-96, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23932718

RESUMEN

Calnexin is a well-characterized transmembrane chaperone involved in the folding of newly synthesized glycoproteins in the lumen of the endoplasmic reticulum (ER). Here, we reveal a previously unrecognized function of calnexin in regulating the transcriptional response downstream of epidermal growth factor receptor (EGF), the product of a well-known human oncogene. We find that cell stimulation with EGF leads to the caspase-8-dependent cleavage of the calnexin cytoplasmic domain, preferentially at ER-mitochondria interaction sites. The released fragment translocates into the nucleus, binds to PIAS3--a natural inhibitor of activated STAT3--and, thus, acts as an enhancer of the STAT3-mediated transcriptional response to EGF. Also, we reveal the unsuspected capacity of calnexin to sense ER stress and, in response, prevent the EGF-induced processing of its cytosolic domain. Thus, cells integrate the health status of the ER to determine the amplitude of their response to EGF.


Asunto(s)
Calnexina/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Receptores ErbB/metabolismo , Factor de Transcripción STAT3/metabolismo , Secuencia de Aminoácidos , Calnexina/química , Caspasa 8/metabolismo , Línea Celular , Receptores ErbB/genética , Humanos , Mitocondrias/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Transducción de Señal , Transcripción Genética
16.
Mol Biol Cell ; 23(14): 2712-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22648169

RESUMEN

Mammalian cells secrete a large number of small proteins, but their mode of translocation into the endoplasmic reticulum is not fully understood. Cotranslational translocation was expected to be inefficient due to the small time window for signal sequence recognition by the signal recognition particle (SRP). Impairing the SRP pathway and reducing cellular levels of the translocon component Sec62 by RNA interference, we found an alternate, Sec62-dependent translocation path in mammalian cells required for the efficient translocation of small proteins with N-terminal signal sequences. The Sec62-dependent translocation occurs posttranslationally via the Sec61 translocon and requires ATP. We classified preproteins into three groups: 1) those that comprise ≤100 amino acids are strongly dependent on Sec62 for efficient translocation; 2) those in the size range of 120-160 amino acids use the SRP pathway, albeit inefficiently, and therefore rely on Sec62 for efficient translocation; and 3) those larger than 160 amino acids depend on the SRP pathway to preserve a transient translocation competence independent of Sec62. Thus, unlike in yeast, the Sec62-dependent translocation pathway in mammalian cells serves mainly as a fail-safe mechanism to ensure efficient secretion of small proteins and provides cells with an opportunity to regulate secretion of small proteins independent of the SRP pathway.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transporte de Proteínas , Proteínas/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Adenosina Trifosfato/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas de Transporte de Membrana/genética , Señales de Clasificación de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal
17.
EMBO J ; 31(7): 1823-35, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22314232

RESUMEN

A third of the human genome encodes N-glycosylated proteins. These are co-translationally translocated into the lumen/membrane of the endoplasmic reticulum (ER) where they fold and assemble before they are transported to their final destination. Here, we show that calnexin, a major ER chaperone involved in glycoprotein folding is palmitoylated and that this modification is mediated by the ER palmitoyltransferase DHHC6. This modification leads to the preferential localization of calnexin to the perinuclear rough ER, at the expense of ER tubules. Moreover, palmitoylation mediates the association of calnexin with the ribosome-translocon complex (RTC) leading to the formation of a supercomplex that recruits the actin cytoskeleton, leading to further stabilization of the assembly. When formation of the calnexin-RTC supercomplex was affected by DHHC6 silencing, mutation of calnexin palmitoylation sites or actin depolymerization, folding of glycoproteins was impaired. Our findings thus show that calnexin is a stable component of the RTC in a manner that is exquisitely dependent on its palmitoylation status. This association is essential for the chaperone to capture its client proteins as they emerge from the translocon, acquire their N-linked glycans and initiate folding.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calnexina/metabolismo , Lipoilación , Glicoproteínas de Membrana/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Péptidos/metabolismo , Ribosomas/metabolismo , Aciltransferasas/genética , Aciltransferasas/metabolismo , Citoesqueleto/metabolismo , Retículo Endoplásmico/metabolismo , Silenciador del Gen , Glicoproteínas/metabolismo , Células HeLa , Humanos , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Estabilidad Proteica
18.
EMBO Mol Med ; 3(4): 208-21, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21328543

RESUMEN

Hyaline Fibromatosis Syndrome (HFS) is a human genetic disease caused by mutations in the anthrax toxin receptor 2 (or cmg2) gene, which encodes a membrane protein thought to be involved in the homeostasis of the extracellular matrix. Little is known about the structure and function of the protein or the genotype­phenotype relationship of the disease. Through the analysis of four patients, we identify three novel mutants and determine their effects at the cellular level. Altogether, we show that missense mutations that map to the extracellular von Willebrand domain or the here characterized Ig-like domain of CMG2 lead to folding defects and thereby to retention of the mutated protein in the endoplasmic reticulum (ER). Mutations in the Ig-like domain prevent proper disulphide bond formation and are more efficiently targeted to ER-associated degradation. Finally, we show that mutant CMG2 can be rescued in fibroblasts of some patients by treatment with proteasome inhibitors and that CMG2 is then properly transported to the plasma membrane and signalling competent, identifying the ER folding and degradation pathway components as promising drug targets for HFS.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Síndrome de Fibromatosis Hialina/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mutación Missense , Inhibidores de Proteasoma , Adolescente , Preescolar , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Síndrome de Fibromatosis Hialina/tratamiento farmacológico , Síndrome de Fibromatosis Hialina/metabolismo , Lactante , Masculino , Proteínas de la Membrana/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores de Péptidos
19.
RNA ; 16(5): 969-79, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20348448

RESUMEN

The signal recognition particle (SRP) is a ubiquitous cytoplasmic ribonucleoprotein complex required for the cotranslational targeting of proteins to the endoplasmic reticulum (ER). In eukaryotes, SRP has to arrest the elongation of the nascent chains during targeting to ensure efficient translocation of the preprotein, and this function of SRP is dependent on SRP9/14. Here we present the results of a mutational study on the human protein h9/14 that identified and characterized regions and single residues essential for elongation arrest activity. Effects of the mutations were assessed both in cell-free translation/translocation assays and in cultured mammalian cells. We identified two patches of basic amino acid residues that are essential for activity, whereas the internal loop of SRP14 was found to be dispensable. One patch of important basic residues comprises the previously identified basic pentapetide KRDKK, which can be substituted by four lysines without loss of function. The other patch includes three lysines in the solvent-accessible alpha2 of h9. All essential residues are located in proximity in SRP9/14 and their basic character suggests that they serve as a positively charged platform for interactions with ribosomal RNA. In addition, they can all be lysines consistent with the hypothesis that they recognize their target(s) via electrostatic contacts, most likely with the phosphate backbone, as opposed to contacts with specific bases.


Asunto(s)
Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Línea Celular , Secuencia Conservada , Prueba de Complementación Genética , Humanos , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Extensión de la Cadena Peptídica de Translación , Multimerización de Proteína , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Partícula de Reconocimiento de Señal/genética , Electricidad Estática
20.
Retrovirology ; 5: 54, 2008 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-18597676

RESUMEN

Human APOBEC3 proteins are editing enzymes that can interfere with the replication of exogenous retroviruses such as human immunodeficiency virus (HIV), hepadnaviruses such as hepatitis B virus (HBV), and with the retrotransposition of endogenous retroelements such as long-interspersed nuclear elements (LINE) and Alu. Here, we show that APOBEC3G, but not other APOBEC3 family members, binds 7SL RNA, the common ancestor of Alu RNAs that is specifically recruited into HIV virions. Our data further indicate that APOBEC3G recognizes 7SL RNA and Alu RNA by its common structure, the Alu domain, suggesting a mechanism for APOBEC3G- mediated inhibition of Alu retrotransposition. However, we also demonstrate that APOBEC3F and APOBEC3G are normally recruited into and inhibit the infectivity of DeltaVif HIV1 virions when 7SLRNA is prevented from accessing particles by RNA interference against SRP14 or by over expression of SRP19, both components of the signal recognition particle. We thus conclude that 7SL RNA is not an essential mediator of the virion packaging of these antiviral cytidine deaminases.


Asunto(s)
Elementos Alu/genética , Citidina Desaminasa/metabolismo , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Desaminasa APOBEC-3G , Elementos Alu/fisiología , Secuencia de Bases , Línea Celular , Citidina Desaminasa/genética , VIH-1/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , ARN Citoplasmático Pequeño/química , Partícula de Reconocimiento de Señal/química , Virión/metabolismo , Ensamble de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...