Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33975959

RESUMEN

Ketamine is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid antidepressant action in some patients with treatment-resistant depression. However, recent data suggest that ∼50% of patients with treatment-resistant depression do not respond to ketamine. The factors that contribute to the nonresponsiveness to ketamine's antidepressant action remain unclear. Recent studies have reported a role for secreted glycoprotein Reelin in regulating pre- and postsynaptic function, which suggests that Reelin may be involved in ketamine's antidepressant action, although the premise has not been tested. Here, we investigated whether the disruption of Reelin-mediated synaptic signaling alters ketamine-triggered synaptic plasticity and behavioral effects. To this end, we used mouse models with genetic deletion of Reelin or apolipoprotein E receptor 2 (Apoer2), as well as pharmacological inhibition of their downstream effectors, Src family kinases (SFKs) or phosphoinositide 3-kinase. We found that disruption of Reelin, Apoer2, or SFKs blocks ketamine-driven behavioral changes and synaptic plasticity in the hippocampal CA1 region. Although ketamine administration did not affect tyrosine phosphorylation of DAB1, an adaptor protein linked to downstream signaling of Reelin, disruption of Apoer2 or SFKs impaired baseline NMDA receptor-mediated neurotransmission. These results suggest that maintenance of baseline NMDA receptor function by Reelin signaling may be a key permissive factor required for ketamine's antidepressant effects. Taken together, our results suggest that impairments in Reelin-Apoer2-SFK pathway components may in part underlie nonresponsiveness to ketamine's antidepressant action.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Ketamina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Proteína Reelina/fisiología , Animales , Proteínas Relacionadas con Receptor de LDL/fisiología , Masculino , Ratones , Plasticidad Neuronal/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/fisiología
2.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-32664652

RESUMEN

Myeloid cells are crucial for the development of vascular inflammation. Low-density lipoprotein receptor-related protein 8 (LRP8) or Apolipoprotein E receptor 2 (ApoER2), is expressed by macrophages, endothelial cells and platelets and has been implicated in the development of cardiovascular diseases. Our aim was to evaluate the role of LRP8, in particular from immune cells, in the development of vascular inflammation. METHODS: LRP8+/+ and LRP8-/- mice (on B6;129S background) were infused with angiotensin II (AngII, 1 mg/kg/day for 7 to 28 day) using osmotic minipumps. Blood pressure was recorded using tail cuff measurements. Vascular reactivity was assessed in isolated aortic segments. Leukocyte activation and infiltration were assessed by flow cytometry of aortic tissue and intravital videomicroscopy imaging. Histological analysis of aortic sections was conducted using sirius red staining. RESULTS: AngII infusion worsened endothelial-dependent vascular relaxation and immune cells rolling and adherence to the carotid artery in both LRP8+/+ as well as LRP8-/- mice. However, only LRP8-/- mice demonstrated a drastically increased mortality rate in response to AngII due to aortic dissection. Bone marrow transplantation revealed that chimeras with LRP8 deficient myeloid cells phenocopied LRP8-/- mice. CONCLUSION: AngII-infused LRP8 deficient mice could be a useful animal model to study aortic dissection reflecting the lethality of this disease in humans.


Asunto(s)
Angiotensina II/toxicidad , Disección Aórtica/inducido químicamente , Proteínas Relacionadas con Receptor de LDL/deficiencia , Acetilcolina/farmacología , Animales , Presión Sanguínea , Trasplante de Médula Ósea , Adhesión Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Bombas de Infusión , Proteínas Relacionadas con Receptor de LDL/fisiología , Masculino , Ratones , Microscopía por Video , Células Mieloides/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Quimera por Radiación , Vasodilatación/efectos de los fármacos
4.
Nat Rev Dis Primers ; 5(1): 30, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31048702

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease caused by antibodies against the acetylcholine receptor (AChR), muscle-specific kinase (MuSK) or other AChR-related proteins in the postsynaptic muscle membrane. Localized or general muscle weakness is the predominant symptom and is induced by the antibodies. Patients are grouped according to the presence of antibodies, symptoms, age at onset and thymus pathology. Diagnosis is straightforward in most patients with typical symptoms and a positive antibody test, although a detailed clinical and neurophysiological examination is important in antibody-negative patients. MG therapy should be ambitious and aim for clinical remission or only mild symptoms with near-normal function and quality of life. Treatment should be based on MG subgroup and includes symptomatic treatment using acetylcholinesterase inhibitors, thymectomy and immunotherapy. Intravenous immunoglobulin and plasma exchange are fast-acting treatments used for disease exacerbations, and intensive care is necessary during exacerbations with respiratory failure. Comorbidity is frequent, particularly in elderly patients. Active physical training should be encouraged.


Asunto(s)
Miastenia Gravis/diagnóstico , Miastenia Gravis/terapia , Acetilcolinesterasa/genética , Acetilcolinesterasa/fisiología , Corticoesteroides/uso terapéutico , Agrina/genética , Agrina/fisiología , Antiinflamatorios no Esteroideos/uso terapéutico , Autoanticuerpos/análisis , Autoanticuerpos/sangre , Biomarcadores/análisis , Biomarcadores/sangre , Blefaroptosis/etiología , Colágeno/genética , Colágeno/fisiología , Cortactina/genética , Cortactina/fisiología , Electromiografía/métodos , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Miastenia Gravis/fisiopatología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/genética , Receptores Colinérgicos/fisiología , Receptores Nicotínicos/genética , Factores de Riesgo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología
5.
Proc Natl Acad Sci U S A ; 114(5): 1159-1164, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28100490

RESUMEN

The kainate receptor (KAR), a subtype of glutamate receptor, mediates excitatory synaptic responses at a subset of glutamatergic synapses. However, the molecular mechanisms underlying the trafficking of its different subunits are poorly understood. Here we use the CA1 hippocampal pyramidal cell, which lacks KAR-mediated synaptic currents, as a null background to determine the minimal requirements for the extrasynaptic and synaptic expression of the GluK2 subunit. We find that the GluK2 receptor itself, in contrast to GluK1, traffics to the neuronal surface and synapse efficiently and the auxiliary subunits Neto1 and Neto2 caused no further enhancement of these two trafficking processes. However, the regulation of GluK2 biophysical properties by Neto proteins is the same as that of GluK1. We further determine that it is the amino-terminal domains (ATDs) of GluK1 and GluK2 that control the strikingly different trafficking properties between these two receptors. Moreover, the ATDs are critical for synaptic expression of heteromeric receptors at mossy fiber-CA3 synapses and also mediate the differential dependence on Neto proteins for surface and synaptic trafficking of GluK1 and GluK2. These results highlight the fundamental differences between the two major KAR subunits and their interplay with Neto auxiliary proteins.


Asunto(s)
Región CA1 Hipocampal/fisiología , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas de la Membrana/fisiología , Transporte de Proteínas/fisiología , Células Piramidales/fisiología , Receptores de Ácido Kaínico/fisiología , Sustitución de Aminoácidos , Animales , Dimerización , Proteínas Relacionadas con Receptor de LDL/química , Proteínas de la Membrana/química , Ratones , Potenciales Postsinápticos Miniatura/fisiología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Dominios Proteicos , Mapas de Interacción de Proteínas , Ratas , Receptores de Ácido Kaínico/química , Receptores de Ácido Kaínico/genética , Receptores de N-Metil-D-Aspartato , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Transmisión Sináptica/fisiología , Receptor de Ácido Kaínico GluK2
6.
Clin Chim Acta ; 457: 137-41, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-27095609

RESUMEN

BACKGROUND: Smooth muscle cell (SMC) migration from the media to the intima, a process affecting plaque stability in advanced-stage atherosclerosis, is under the control of LR11. To delineate the clinical significance of the circulating soluble form of LR11 (sLR11) in patients with type 2 diabetes (T2D), we analyzed the correlation of sLR11 levels with intima-media thickness (IMT) of carotid arteries. METHODS: Plasma sLR11 levels were measured in 165 patients with T2D (mean age 56.2±10.4 y, 58.2% males, and BMI 24.6±3.6) by ELISA. Averaged IMT levels of common carotid arteries were determined by ultrasonography. RESULTS: Circulating sLR11 levels were 9.8±3.5ng/ml, and correlated positively with the classical atherosclerosis risk factors age, sex, systolic blood pressure, low-density lipoprotein-cholesterol (LDL-C), fasting plasma-glucose (FPG), and glycosylated hemoglobin. Multivariate linear regression analysis indicated that only FPG was associated with sLR11; sLR11 correlated positively with IMT, together with age and FPG, but less with LDL-C. Among the serum risk factors for IMT, multivariate linear regression analysis uncovered that sLR11 was independently associated with IMT. Subsequent logistic analysis revealed that FPG correlated best with IMT values at a cut-off of 0.80mm and sLR11 at a cut-off of 0.90mm, respectively, while LDL-C showed lower discriminatory power at any IMT cut-off values. CONCLUSION: Increased sLR11 concentrations are highly associated with increased IMT as well as with FPG in middle-aged, non-obese patients with T2D. Circulating sLR11 may be a novel marker representing the pathophysiology of intimal SMCs in patients with T2D.


Asunto(s)
Biomarcadores/sangre , Arterias Carótidas/patología , Movimiento Celular/fisiología , Diabetes Mellitus Tipo 2/patología , Proteínas Relacionadas con Receptor de LDL/sangre , Proteínas de Transporte de Membrana/sangre , Músculo Liso Vascular/patología , Túnica Íntima/patología , Anciano , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Proteínas Relacionadas con Receptor de LDL/fisiología , Masculino , Proteínas de Transporte de Membrana/fisiología , Persona de Mediana Edad , Estudios Prospectivos
7.
Front Med ; 10(1): 76-84, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26620053

RESUMEN

Anti-ß2 glycoprotein I (anti-ß2GPI ) antibodies are important contributors to thrombosis, especially in patients with antiphospholipid syndrome (APS). However, the mechanism by which anti-ß2GPI antibodies are involved in the pathogenesis of thrombosis is not fully understood. In this report, we investigated the role of anti- ß2GPI antibodies in complexes with ß2GPI as mediators of platelet activation, which can serve as a potential source contributing to thrombosis. We examined the involvement of the apolipoprotein E receptor 2' (apoER2') and glycoprotein I ba (GP I ba) in platelet activation induced by the anti-ß2GPI /ß2GPI complex. The interaction between the anti-ß2GPI /ß2GPI complex and platelets was examined using in vitro methods, in which the Fc portion of the antibody was immobilized using protein A coated onto a microtiter plate. Platelet activation was assessed by measuring GPII b/III a activation and P-selectin expression and thromboxane B2 production as well as p38 mitogen-activated protein kinase phosphorylation. Our results revealed that the anti-ß2GPI /ß2GPI complex was able to activate platelets, and this activation was inhibited by either the anti-GP I bα antibody or the apoER2' inhibitor. Results showed that the anti-ß2GPI /ß2GPI complex induced platelet activation via GPI ba and apoER2', which may then contribute to the prothrombotic tendency in APS patients.


Asunto(s)
Autoanticuerpos/inmunología , Glicoproteínas/fisiología , Inmunoglobulinas/fisiología , Proteínas Relacionadas con Receptor de LDL/fisiología , Activación Plaquetaria/fisiología , beta 2 Glicoproteína I/inmunología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , beta 2 Glicoproteína I/fisiología
8.
Brain Struct Funct ; 221(8): 4103-4110, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26627482

RESUMEN

The sortilin-related receptor 1 (SORL1) gene has been associated with late-onset Alzheimer's disease (LOAD) and structural impairments in several ethnic populations. However, how this gene affects brain function properties remains unclear. We investigated associations of SORL1 rs2070045 with functional connectivity density (FCD) in healthy young adults. This single-nucleotide polymorphism was selected because it is the SORL1 variant that has been frequently associated with LOAD in several populations, including the Chinese Han population. A total of 275 healthy young Chinese Han subjects with successful genotyping and MRI examinations were included. The effect of SORL1 rs2070045 was explored using a voxel-wise FCD analysis. A significant effect of SORL1 rs2070045 on the FCD was found in the right inferior temporal gyrus. The risk G allele carriers of the rs2070045 exhibited a lower FCD than the protective TT carriers. This effect was independent of the status of apolipoprotein E. This study provides the first evidence that the SORL1 gene is associated with brain FCD differences in healthy young adults. In the genetic risk subjects, connectivity impairment already starts during young adulthood, which may predispose the risk allele carriers to be susceptible to LOAD after several decades.


Asunto(s)
Encéfalo/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/fisiología , Adolescente , Adulto , Pueblo Asiatico , Mapeo Encefálico , Femenino , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Memoria/fisiología , Polimorfismo de Nucleótido Simple , Adulto Joven
9.
Ann Rheum Dis ; 75(6): 1228-35, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26290588

RESUMEN

OBJECTIVES: Osteoarthritis (OA) is a leading cause of disability for which there is no cure. The identification of molecules supporting cartilage homeostasis and regeneration is therefore a major pursuit in musculoskeletal medicine. Agrin is a heparan sulfate proteoglycan which, through binding to low-density lipoprotein receptor-related protein 4 (LRP4), is required for neuromuscular synapse formation. In other tissues, it connects the cytoskeleton to the basement membrane through binding to α-dystroglycan. Prompted by an unexpected expression pattern, we investigated the role and receptor usage of agrin in cartilage. METHODS: Agrin expression pattern was investigated in human osteoarthritic cartilage and following destabilisation of the medial meniscus in mice. Extracellular matrix (ECM) formation and chondrocyte differentiation was studied in gain and loss of function experiments in vitro in three-dimensional cultures and gain of function in vivo, using an ectopic cartilage formation assay in nude mice. Receptor usage was investigated by disrupting LRP4 and α-dystroglycan by siRNA and blocking antibodies respectively. RESULTS: Agrin was detected in normal cartilage but was progressively lost in OA. In vitro, agrin knockdown resulted in reduced glycosaminoglycan content, downregulation of the cartilage transcription factor SOX9 and other cartilage-specific ECM molecules. Conversely, exogenous agrin supported cartilage differentiation in vitro and ectopic cartilage formation in vivo. In the context of cartilage differentiation, agrin used an unusual receptor repertoire requiring both LRP4 and α-dystroglycan. CONCLUSIONS: We have discovered that agrin strongly promotes chondrocyte differentiation and cartilage formation in vivo. Our results identify agrin as a novel potent anabolic growth factor with strong therapeutic potential in cartilage regeneration.


Asunto(s)
Agrina/fisiología , Artritis Experimental/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Distroglicanos/fisiología , Osteoartritis/metabolismo , Receptores de LDL/fisiología , Agrina/biosíntesis , Agrina/genética , Agrina/farmacología , Animales , Artritis Experimental/genética , Artritis Experimental/patología , Cartílago Articular/patología , Células Cultivadas , Condrogénesis/efectos de los fármacos , Regulación hacia Abajo/fisiología , Técnicas de Silenciamiento del Gen , Homeostasis/fisiología , Humanos , Proteínas Relacionadas con Receptor de LDL/fisiología , Masculino , Ratones Endogámicos DBA , Ratones Noqueados , Osteoartritis/genética , Osteoartritis/patología , Osteogénesis/fisiología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Factor de Transcripción SOX9/biosíntesis , Factor de Transcripción SOX9/genética , Regulación hacia Arriba/fisiología
10.
J Neurosci ; 35(37): 12703-13, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377460

RESUMEN

SORLA is a neuronal sorting receptor implicated both in sporadic and familial forms of AD. SORLA reduces the amyloidogenic burden by two mechanisms, either by rerouting internalized APP molecules from endosomes to the trans-Golgi network (TGN) to prevent proteolytic processing or by directing newly produced Aß to lysosomes for catabolism. Studies in cell lines suggested that the interaction of SORLA with cytosolic adaptors retromer and GGA is required for receptor sorting to and from the TGN. However, the relevance of anterograde or retrograde trafficking for SORLA activity in vivo remained largely unexplored. Here, we generated mouse models expressing SORLA variants lacking binding sites for GGA or retromer to query this concept in the brain. Disruption of retromer binding resulted in a retrograde-sorting defect with accumulation of SORLA in endosomes and depletion from the TGN, and in an overall enhanced APP processing. In contrast, disruption of the GGA interaction did not impact APP processing but caused increased brain Aß levels, a mechanism attributed to a defect in anterograde lysosomal targeting of Aß. Our findings substantiated the significance of adaptor-mediated sorting for SORLA activities in vivo, and they uncovered that anterograde and retrograde sorting paths may serve discrete receptor functions in amyloidogenic processes. SIGNIFICANCE STATEMENT: SORLA is a sorting receptor that directs target proteins to distinct intracellular compartments in neurons. SORLA has been identified as a genetic risk factor for sporadic, but recently also for familial forms of AD. To confirm the relevance of SORLA sorting for AD processes in the brain, we generated mouse lines, which express trafficking mutants instead of the wild-type form of this receptor. Studying neuronal activities in these mutant mice, we dissected distinct trafficking routes for SORLA guided by two cytosolic adaptors termed GGA and retromer. We show that these sorting pathways serve discrete functions in control of amyloidogenic processes and may represent unique therapeutic targets to interfere with specific aspects of neurodegenerative processes in the diseased brain.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas de Transporte de Membrana/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Péptidos beta-Amiloides/metabolismo , Animales , Sitios de Unión , Línea Celular , Endosomas/metabolismo , Femenino , Hipocampo/citología , Proteínas Relacionadas con Receptor de LDL/metabolismo , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , ARN no Traducido/genética , Proteínas Recombinantes de Fusión/metabolismo , Red trans-Golgi/metabolismo
11.
Rev Endocr Metab Disord ; 16(2): 141-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26048454

RESUMEN

A dozen years ago the identification of causal mutations in the low-density lipoprotein receptor-related protein 5 (LRP5) gene involved in two rare bone disorders propelled research in the bone field in totally new directions. Since then, there have been an explosion in the number of reports that highlight the role of the Wnt/ß-catenin pathway in the regulation of bone homeostasis. In this review we discuss some of the most recent reports (in the past 2 years) highlighting the involvement of the members of the LRP family (LRP5, LRP6, LRP4, and more recently LRP8) in the maintenance of bone and their implications in bone diseases. These reports include records of new single nucleotides polymorphisms (SNPs) and haplotypes that suggest variants in these genes can contribute to subtle variation in bone traits to mutations that give rise to extreme bone phenotypes. All of these serve to further support and reinforce the importance of this tightly regulated pathway in bone. Furthermore, we discuss provocative reports suggesting novel approaches through inhibitors of this pathway to treat rarer diseases such as Osteoporosis-Pseudoglioma Syndrome (OPPG), Osteogenesis Imperfecta (OI), and Sclerosteosis/Van Buchem disease. It is hoped that by understanding the role of each component of the pathway and their involvement in bone diseases that this knowledge will allow us to develop new, more effective therapeutic approaches for more common diseases such as post-menopausal osteoporosis, osteoarthritis, and rheumatoid arthritis as well as these rarer bone diseases.


Asunto(s)
Enfermedades Óseas/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/terapia , Enfermedades Óseas/terapia , Humanos , Proteínas Relacionadas con Receptor de LDL/genética , Familia de Multigenes/fisiología , Mutación/fisiología , Osteoartritis/genética , Osteoartritis/terapia , Osteoporosis/genética , Osteoporosis/terapia , Vía de Señalización Wnt/genética
12.
Annu Rev Nutr ; 35: 109-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25974694

RESUMEN

Selenium is regulated in the body to maintain vital selenoproteins and to avoid toxicity. When selenium is limiting, cells utilize it to synthesize the selenoproteins most important to them, creating a selenoprotein hierarchy in the cell. The liver is the central organ for selenium regulation and produces excretory selenium forms to regulate whole-body selenium. It responds to selenium deficiency by curtailing excretion and secreting selenoprotein P (Sepp1) into the plasma at the expense of its intracellular selenoproteins. Plasma Sepp1 is distributed to tissues in relation to their expression of the Sepp1 receptor apolipoprotein E receptor-2, creating a tissue selenium hierarchy. N-terminal Sepp1 forms are taken up in the renal proximal tubule by another receptor, megalin. Thus, the regulated whole-body pool of selenium is shifted to needy cells and then to vital selenoproteins in them to supply selenium where it is needed, creating a whole-body selenoprotein hierarchy.


Asunto(s)
Homeostasis/fisiología , Selenio/metabolismo , Animales , Disponibilidad Biológica , Transporte Biológico , Biomarcadores , Dieta , Suplementos Dietéticos , Estado de Salud , Humanos , Túbulos Renales Proximales/metabolismo , Proteínas Relacionadas con Receptor de LDL/fisiología , Hígado/fisiología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Necesidades Nutricionales , Especificidad de Órganos , Selenio/deficiencia , Selenio/farmacocinética , Selenocisteína/metabolismo , Selenometionina/metabolismo , Selenoproteína P/análisis , Selenoproteína P/sangre , Selenoproteínas/biosíntesis , Selenoproteínas/metabolismo
13.
Nat Struct Mol Biol ; 22(3): 199-206, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25643321

RESUMEN

SorLA is a neuronal sorting receptor considered to be a major risk factor for Alzheimer's disease. We have recently reported that it directs lysosomal targeting of nascent neurotoxic amyloid-ß (Aß) peptides by directly binding Aß. Here, we determined the crystal structure of the human sorLA domain responsible for Aß capture, Vps10p, in an unbound state and in complex with two ligands. Vps10p assumes a ten-bladed ß-propeller fold with a large tunnel at the center. An internal ligand derived from the sorLA propeptide bound inside the tunnel to extend the ß-sheet of one of the propeller blades. The structure of the sorLA Vps10p-Aß complex revealed that the same site is used. Peptides are recognized by sorLA Vps10p in redundant modes without strict dependence on a particular amino acid sequence, thus suggesting a broad specificity toward peptides with a propensity for ß-sheet formation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas Relacionadas con Receptor de LDL/química , Proteínas de Transporte de Membrana/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/fisiología , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína
16.
Sci Signal ; 7(353): ra113, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25429077

RESUMEN

Apoer2 is an essential receptor in the central nervous system that binds to the apolipoprotein ApoE. Various splice variants of Apoer2 are produced. We showed that Apoer2 lacking exon 16, which encodes the O-linked sugar (OLS) domain, altered the proteolytic processing and abundance of Apoer2 in cells and synapse number and function in mice. In cultured cells expressing this splice variant, extracellular cleavage of OLS-deficient Apoer2 was reduced, consequently preventing γ-secretase-dependent release of the intracellular domain of Apoer2. Mice expressing Apoer2 lacking the OLS domain had increased Apoer2 abundance in the brain, hippocampal spine density, and glutamate receptor abundance, but decreased synaptic efficacy. Mice expressing a form of Apoer2 lacking the OLS domain and containing an alternatively spliced cytoplasmic tail region that promotes glutamate receptor signaling showed enhanced hippocampal long-term potentiation (LTP), a phenomenon associated with learning and memory. However, these mice did not display enhanced spatial learning in the Morris water maze, and cued fear conditioning was reduced. Reducing the expression of the mutant Apoer2 allele so that the abundance of the protein was similar to that of Apoer2 in wild-type mice normalized spine density, hippocampal LTP, and cued fear learning. These findings demonstrated a role for ApoE receptors as regulators of synaptic glutamate receptor activity and established differential receptor glycosylation as a potential regulator of synaptic function and memory.


Asunto(s)
Empalme Alternativo , Reacción de Prevención/fisiología , Región CA1 Hipocampal/fisiopatología , Miedo/fisiología , Proteínas Relacionadas con Receptor de LDL/fisiología , Potenciación a Largo Plazo/fisiología , Aprendizaje por Laberinto/fisiología , Proteínas del Tejido Nervioso/fisiología , Procesamiento Proteico-Postraduccional , Transmisión Sináptica/fisiología , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Región CA1 Hipocampal/metabolismo , Condicionamiento Clásico/fisiología , Señales (Psicología) , Dendritas/ultraestructura , Exones , Femenino , Glicosilación , Proteínas Relacionadas con Receptor de LDL/química , Proteínas Relacionadas con Receptor de LDL/genética , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Reflejo de Sobresalto/fisiología , Relación Estructura-Actividad , Transmisión Sináptica/genética
17.
Biochim Biophys Acta ; 1842(9): 1395-405, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24840660

RESUMEN

Genome-wide association studies have linked LRP8 polymorphisms to premature coronary artery disease and myocardial infarction in humans. However, the mechanisms by which dysfunctions of apolipoprotein E receptor-2 (apoER2), the protein encoded by LRP8 gene, influence atherosclerosis have not been elucidated completely. The current study focused on the role of apoER2 in macrophages, a cell type that plays an important role in atherosclerosis. Results showed that apoER2-deficient mouse macrophages accumulated more lipids and were more susceptible to oxidized LDL (oxLDL)-induced death compared to control cells. Consistent with these findings, apoER2 deficient macrophages also displayed defective serum-induced Akt activation and higher levels of the pro-apoptotic protein phosphorylated p53. Furthermore, the expression and activation of peroxisome proliferator-activated receptor γ (PPARγ) were increased in apoER2-deficient macrophages. Deficiency of apoER2 in hypercholesterolemic LDL receptor-null mice (Lrp8(-/-)Ldlr(-/-) mice) also resulted in accelerated atherosclerosis with more complex lesions and extensive lesion necrosis compared to Lrp8(+/+)Ldlr(-/-) mice. The atherosclerotic plaques of Lrp8(-/-)Ldlr(-/-) mice displayed significantly higher levels of p53-positive macrophages, indicating that the apoER2-deficient macrophages contribute to the accelerated atherosclerotic lesion necrosis observed in these animals. Taken together, this study indicates that apoER2 in macrophages limits PPARγ expression and protects against oxLDL-induced cell death. Thus, abnormal apoER2 functions in macrophages may at least in part contribute to the premature coronary artery disease and myocardial infarction in humans with LRP8 polymorphisms. Moreover, the elevated PPARγ expression in apoER2-deficient macrophages suggests that LRP8 polymorphism may be a genetic modifier of cardiovascular risk with PPARγ therapy.


Asunto(s)
Hipercolesterolemia/patología , Proteínas Relacionadas con Receptor de LDL/fisiología , Macrófagos/patología , PPAR gamma/metabolismo , Placa Aterosclerótica/patología , Receptores de LDL/fisiología , Animales , Apoptosis , Western Blotting , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Hipercolesterolemia/complicaciones , Hipercolesterolemia/metabolismo , Lípidos/análisis , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Placa Aterosclerótica/etiología , Placa Aterosclerótica/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo
18.
J Biol Chem ; 289(23): 15894-903, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24755222

RESUMEN

ApoE Receptor 2 (ApoER2) and the very low density lipoprotein receptor (VLDLR) are type I transmembrane proteins belonging to the LDLR family of receptors. They are neuronal proteins found in synaptic compartments that play an important role in neuronal migration during development. ApoER2 and VLDLR bind to extracellular glycoproteins, such as Reelin and F-spondin, which leads to phosphorylation of adaptor proteins and subsequent activation of downstream signaling pathways. It is thought that ApoER2 and VLDLR undergo clustering upon binding to their ligands, but no direct evidence of clustering has been shown. Here we show strong clustering of ApoER2 induced by the dimeric ligands Fc-RAP, F-spondin, and Reelin but relatively weak clustering with the ligand apoE in the absence of lipoproteins. This clustering involves numerous proteins besides ApoER2, including amyloid precursor protein and the synaptic adaptor protein PSD-95. Interestingly, we did not observe strong clustering of ApoER2 with VLDLR. Clustering was modulated by both extracellular and intracellular domains of ApoER2. Together, our data demonstrate that several multivalent ligands for ApoER2 induce clustering in transfected cells and primary neurons and that these complexes included other synaptic molecules, such as APP and PSD-95.


Asunto(s)
Proteínas Relacionadas con Receptor de LDL/metabolismo , Animales , Células COS , Movimiento Celular , Chlorocebus aethiops , Proteínas Relacionadas con Receptor de LDL/fisiología , Ligandos , Ratones , Neuronas/fisiología , Fosforilación , Proteína Reelina
19.
FASEB J ; 28(8): 3579-88, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24760755

RESUMEN

Selenoprotein P (Sepp1) and its receptor, apolipoprotein E receptor 2 (apoER2), account for brain retaining selenium better than other tissues. The primary sources of Sepp1 in plasma and brain are hepatocytes and astrocytes, respectively. ApoER2 is expressed in varying amounts by tissues; within the brain it is expressed primarily by neurons. Knockout of Sepp1 or apoER2 lowers brain selenium from ∼120 to ∼50 ng/g and leads to severe neurodegeneration and death in mild selenium deficiency. Interactions of Sepp1 and apoER2 that protect against this injury have not been characterized. We studied Sepp1, apoER2, and brain selenium in knockout mice. Immunocytochemistry showed that apoER2 mediates Sepp1 uptake at the blood-brain barrier. When Sepp1(-/-) or apoER2(-/-) mice developed severe neurodegeneration caused by mild selenium deficiency, brain selenium was ∼35 ng/g. In extreme selenium deficiency, however, brain selenium of ∼12 ng/g was tolerated when both Sepp1 and apoER2 were intact in the brain. These findings indicate that tandem Sepp1-apoER2 interactions supply selenium for maintenance of brain neurons. One interaction is at the blood-brain barrier, and the other is within the brain. We postulate that Sepp1 inside the blood-brain barrier is taken up by neurons via apoER2, concentrating brain selenium in them.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/fisiología , Degeneración Nerviosa/prevención & control , Selenio/metabolismo , Selenoproteína P/fisiología , Animales , Animales Congénicos , Transporte Biológico , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Capilares/metabolismo , Plexo Coroideo/embriología , Plexo Coroideo/crecimiento & desarrollo , Plexo Coroideo/metabolismo , Endocitosis , Células Endoteliales/metabolismo , Femenino , Proteínas Relacionadas con Receptor de LDL/deficiencia , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Embarazo , Selenio/administración & dosificación , Selenio/deficiencia , Selenio/farmacocinética , Selenoproteína P/deficiencia
20.
Br Poult Sci ; 54(5): 567-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24286503

RESUMEN

1. Two splice variants of duck LRP8 were identified, one containing 8 ligand-binding repeats (LRP8-1) and the other containing only 7 repeats (LRP8-2). The two transcripts share ~71-91% nucleic acid identity and ~65-94% amino acid identity with their counterparts in other species. A phylogenetic tree based on amino acid sequences shows that duck LRP8 proteins are closely related to those of chicken, turkey and zebra finch. 2. The semi-quantitative reverse transcription polymerase chain reaction (RT-PCR )analysis indicates that the two transcripts are expressed in all the examined tissues, and the LRP8-1 transcript is more highly expressed in hypothalamus, ovary and pituitary gland than in other detected tissues. 3. Six single nucleotide polymorphisms (SNPs) were identified in the coding region. Association analysis demonstrated that the c.528C > T genotypes were associated with egg production (EP) (EP210d, EP300d and EP360d), age at laying the first egg (AFE) and body weight at sexual maturity (BWSM). The c.1371A > G genotypes were associated with egg production (EP210d, EP300d and EP360d). 4. The haplotypes of c.528C > T and c.1371A > G were associated with EP (EP210d, EP300d and EP360d), yolk weight (YW), albumen weight (AW), egg weight (EW), BWSM and the first egg weight (FEW). 5. Duck LRP8 gene was associated with some reproductive traits and is an important candidate gene for the genetic selection of improved reproductive traits.


Asunto(s)
Patos/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas Relacionadas con Receptor de LDL/fisiología , Filogenia , Polimorfismo Conformacional Retorcido-Simple/fisiología , Reproducción/fisiología , Animales , Secuencia de Bases , Clonación Molecular , Cruzamientos Genéticos , Patos/genética , Femenino , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Haplotipos , Proteínas Relacionadas con Receptor de LDL/genética , Datos de Secuencia Molecular , Polimorfismo de Longitud del Fragmento de Restricción , Polimorfismo Conformacional Retorcido-Simple/genética , ARN/química , ARN/genética , Reproducción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA