Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 13(7)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38607022

RESUMEN

Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.


Asunto(s)
Antiinflamatorios , Encefalomielitis Autoinmune Experimental , Proteína Reelina , Animales , Ratones , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Proteína Reelina/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antiinflamatorios/uso terapéutico
2.
Front Immunol ; 14: 1185748, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37441066

RESUMEN

Thromboembolic complications and excessive inflammation are frequent in severe COVID-19, potentially leading to long COVID. In non-COVID studies, we and others demonstrated that circulating Reelin promotes leukocyte infiltration and thrombosis. Thus, we hypothesized that Reelin participates in endothelial dysfunction and hyperinflammation during COVID-19. We showed that Reelin was increased in COVID-19 patients and correlated with the disease activity. In the severe COVID-19 group, we observed a hyperinflammatory state, as judged by increased concentration of cytokines (IL-1α, IL-4, IL-6, IL-10 and IL-17A), chemokines (IP-10 and MIP-1ß), and adhesion markers (E-selectin and ICAM-1). Reelin level was correlated with IL-1α, IL-4, IP-10, MIP-1ß, and ICAM-1, suggesting a specific role for Reelin in COVID-19 progression. Furthermore, Reelin and all of the inflammatory markers aforementioned returned to normal in a long COVID cohort, showing that the hyperinflammatory state was resolved. Finally, we tested Reelin inhibition with the anti-Reelin antibody CR-50 in hACE2 transgenic mice infected with SARS-CoV-2. CR-50 prophylactic treatment decreased mortality and disease severity in this model. These results demonstrate a direct proinflammatory function for Reelin in COVID-19 and identify it as a drug target. This work opens translational clinical applications in severe SARS-CoV-2 infection and beyond in auto-inflammatory diseases.


Asunto(s)
COVID-19 , Ratones , Animales , Humanos , Molécula 1 de Adhesión Intercelular , Síndrome Post Agudo de COVID-19 , SARS-CoV-2 , Quimiocina CCL4 , Quimiocina CXCL10 , Interleucina-4 , Inflamación
3.
Sci Immunol ; 6(62)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34452924

RESUMEN

Under normal conditions, the blood-brain barrier effectively regulates the passage of immune cells into the central nervous system (CNS). However, under pathological conditions such as multiple sclerosis (MS), leukocytes, especially monocytes, infiltrate the CNS where they promote inflammatory demyelination, resulting in paralysis. Therapies targeting the immune cells directly and preventing leukocyte infiltration exist for MS but may compromise the immune system. Here, we explore how apolipoprotein E receptor 2 (ApoER2) regulates vascular adhesion and infiltration of monocytes during inflammation. We induced experimental autoimmune encephalitis in ApoER2 knockout mice and in mice carrying a loss-of-function mutation in the ApoER2 cytoplasmic domain. In both models, paralysis and neuroinflammation were largely abolished as a result of greatly diminished monocyte adherence due to reduced expression of adhesion molecules on the endothelial surface. Our findings expand our mechanistic understanding of the vascular barrier, the regulation of inflammation and vascular permeability, and the therapeutic potential of ApoER2-targeted therapies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Endotelio Vascular/inmunología , Proteínas Relacionadas con Receptor de LDL/inmunología , Monocitos/inmunología , Animales , Adhesión Celular/inmunología , Proteínas Relacionadas con Receptor de LDL/deficiencia , Masculino , Ratones , Ratones Noqueados
4.
Arterioscler Thromb Vasc Biol ; 41(4): 1309-1318, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626909

RESUMEN

[Figure: see text].


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Aterosclerosis/prevención & control , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Adhesión Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Rodamiento de Leucocito/efectos de los fármacos , Leucocitos/efectos de los fármacos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Masculino , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Proteína Reelina , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Transducción de Señal , Células U937
5.
Sci Transl Med ; 12(556)2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32801146

RESUMEN

Neuroinflammation as a result of immune cell recruitment into the central nervous system (CNS) is a key pathogenic mechanism of multiple sclerosis (MS). However, current anti-inflammatory interventions depleting immune cells or directly targeting their trafficking into the CNS can have serious side effects, highlighting a need for better immunomodulatory strategies. We detected increased Reelin concentrations in the serum of patients with MS, resulting in increased endothelial permeability to leukocytes through increased nuclear factor κB-mediated expression of vascular adhesion molecules. We thus investigated the prophylactic and therapeutic potential of Reelin immunodepletion in experimental autoimmune encephalomyelitis (EAE) and further validated the results in Reelin knockout mice. Removal of plasma Reelin by either approach protected against neuroinflammation and largely abolished the neurological consequences by reducing endothelial permeability and immune cell accumulation in the CNS. Our findings suggest Reelin depletion as a therapeutic approach with an inherent good safety margin for the treatment of MS and other diseases where leukocyte extravasation is a major driver of pathogenicity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Encefalomielitis , Esclerosis Múltiple , Animales , Sistema Nervioso Central , Humanos , Leucocitos , Ratones , Ratones Endogámicos C57BL , Proteína Reelina
6.
J Neuroinflammation ; 16(1): 257, 2019 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-31810478

RESUMEN

BACKGROUND: Astrocytes contribute to the crosstalk that generates chronic neuro-inflammation in neurological diseases; however, compared with microglia, astrocytes respond to a more limited continuum of innate immune system stimulants. Recent studies suggest that the fibrinolysis system may regulate inflammation. The goal of this study was to test whether fibrinolysis system components activate astrocytes and if so, elucidate the responsible biochemical pathway. METHODS: Primary cultures of astrocytes and microglia were prepared from neonatal mouse brains. The ability of purified fibrinolysis system proteins to elicit a pro-inflammatory response was determined by measuring expression of the mRNAs encoding tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and chemokine (C-C motif) ligand 2 (CCL2). IκBα phosphorylation also was measured. Plasminogen activation in association with cells was detected by chromogenic substrate hydrolysis. The activity of specific receptors was tested using neutralizing antibodies and reagents. RESULTS: Astrocytes expressed pro-inflammatory cytokines when treated with plasminogen but not when treated with agonists for Toll-like Receptor-4 (TLR4), TLR2, or TLR9. Microglia also expressed pro-inflammatory cytokines in response to plasminogen; however, in these cells, the response was observed only when tissue-type plasminogen activator (tPA) was added to activate plasminogen. In astrocytes, endogenously produced urokinase-type plasminogen activator (uPA) converted plasminogen into plasmin in the absence of tPA. Plasminogen activation was dependent on the plasminogen receptor, α-enolase, and the uPA receptor, uPAR. Although uPAR is capable of directly activating cell-signaling, the receptor responsible for cytokine expression and IκBα phosphorylation response to plasmin was Protease-activated Receptor-1 (PAR-1). The pathway, by which plasminogen induced astrocyte activation, was blocked by inhibiting any one of the three receptors implicated in this pathway with reagents such as εACA, α-enolase-specific antibody, uPAR-specific antibody, the uPA amino terminal fragment, or a pharmacologic PAR-1 inhibitor. CONCLUSIONS: Plasminogen may activate astrocytes for pro-inflammatory cytokine expression through the concerted action of at least three distinct fibrinolysis protease receptors. The pathway is dependent on uPA to activate plasminogen, which is expressed endogenously by astrocytes in culture but also may be provided by other cells in the astrocytic cell microenvironment in the CNS.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Citocinas/biosíntesis , Fibrinólisis/fisiología , Fibrinolíticos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Células Cultivadas , Citocinas/genética , Fibrinólisis/efectos de los fármacos , Expresión Génica , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Plasminógeno/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirroles/farmacología , Quinazolinas/farmacología
7.
Alzheimers Dement (N Y) ; 5: 458-467, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921961

RESUMEN

INTRODUCTION: Currently, there is no cure for Alzheimer's disease (AD), and it is widely accepted that AD is a complex disease with multiple approaches necessary to prevent and treat the disease. METHODS: Using amyloid biomarkers in human cerebrospinal fluid, pharmacokinetic, safety, and metabolism studies, we investigate the properties of NGP 555, γ-secretase modulator, for the first time in human clinical trials. RESULTS: Our preclinical and clinical studies combined show beneficial effects with NGP 555 on synaptic response and amyloid cerebrospinal fluid biomarkers while avoiding negative side effects. Importantly, pharmacokinetic and pharmacodynamic parameters combined with safety outcomes indicate that NGP 555 penetrates the blood-brain barrier and increases the ratio of amyloid-ß peptide Aß37 and Aß38 compared with that of Aß42, establishing a proof of target engagement in humans in a 14 day, once-daily oral dosing trial. DISCUSSION: In humans, NGP 555 has demonstrated a beneficial shift in the production of Aß37 and Aß38 versus Aß42 biomarker levels in the cerebrospinal fluid while maintaining an adequate safety profile. The overall clinical goal is to achieve an optimal balance of efficacy for altering amyloid-ß peptide (Aß) biomarkers while maintaining a safe profile so that NGP 555 can be given early in AD to prevent production of Aß42 and accumulation of amyloid plaques, in an effort to prevent aggregation of tau and destruction of neurons and synapses resulting in cognitive decline.

8.
Alzheimers Dement (N Y) ; 3(1): 65-73, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28497107

RESUMEN

INTRODUCTION: Alzheimer's disease (AD) is defined by the progressive accumulation of amyloid plaques and neurofibrillary tangles in the brain which precedes cognitive decline by years. METHODS: Using amyloid biomarkers, chemical modeling, mouse behavioral models, and drug development techniques we investigate the properties of NGP 555, a clinical-stage γ-secretase modulator. RESULTS: NGP 555 shifts amyloid peptide production to the smaller, non-aggregating forms of amyloid. Our preclinical studies show beneficial effects on amyloid biomarkers, pathology, and cognition. NGP 555 has successfully completed chemistry, pharmacology, toxicity, metabolism, and safety studies. DISCUSSION: Abundant data support Aß42 as a target for prophylactic or early-stage intervention therapies in AD. The γ-secretase modulator, NGP 555 is being actively developed in human clinical trials for the prevention of Alzheimer's disease with the overall aim to achieve an appropriate balance of potency/efficacy on reducing the toxic forms of amyloid versus safety.

9.
J Biol Chem ; 285(49): 38042-52, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20921220

RESUMEN

The γ-secretase protein complex executes the intramembrane proteolysis of amyloid precursor protein (APP), which releases Alzheimer disease ß-amyloid peptide. In addition to APP, γ-secretase also cleaves several other type I membrane protein substrates including Notch1 and N-cadherin. γ-Secretase is made of four integral transmembrane protein subunits: presenilin (PS), nicastrin, APH1, and PEN2. Multiple lines of evidence indicate that a heteromer of PS-derived N- and C-terminal fragments functions as the catalytic subunit of γ-secretase. Only limited information is available on the domains within each subunit involved in the recognition and recruitment of diverse substrates and the transfer of substrates to the catalytic site. Here, we performed mutagenesis of two domains of PS1, namely the first luminal loop domain (LL1) and the second transmembrane domain (TM2), and analyzed PS1 endoproteolysis as well as the catalytic activities of PS1 toward APP, Notch, and N-cadherin. Our results show that distinct residues within LL1 and TM2 domains as well as the length of the LL1 domain are critical for PS1 endoproteolysis, but not for PS1 complex formation with nicastrin, APH1, and PEN2. Furthermore, our experimental PS1 mutants formed γ-secretase complexes with distinct catalytic properties toward the three substrates examined in this study; however, the mutations did not affect PS1 interaction with the substrates. We conclude that the N-terminal LL1 and TM2 domains are critical for PS1 endoproteolysis and the coordination between the putative substrate-docking site and the catalytic core of the γ-secretase.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Presenilina-1/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide , Animales , Cadherinas/genética , Cadherinas/metabolismo , Dominio Catalítico/fisiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Mutagénesis , Mutación , Presenilina-1/genética , Estructura Terciaria de Proteína , Receptor Notch1/genética , Receptor Notch1/metabolismo , Especificidad por Sustrato/fisiología
10.
Neuron ; 67(5): 769-80, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20826309

RESUMEN

Alzheimer's disease (AD) is characterized pathologically by the abundance of senile plaques and neurofibrillary tangles in the brain. We synthesized over 1200 novel gamma-secretase modulator (GSM) compounds that reduced Abeta(42) levels without inhibiting epsilon-site cleavage of APP and Notch, the generation of the APP and Notch intracellular domains, respectively. These compounds also reduced Abeta(40) levels while concomitantly elevating levels of Abeta(38) and Abeta(37). Immobilization of a potent GSM onto an agarose matrix quantitatively recovered Pen-2 and to a lesser degree PS-1 NTFs from cellular extracts. Moreover, oral administration (once daily) of another potent GSM to Tg 2576 transgenic AD mice displayed dose-responsive lowering of plasma and brain Abeta(42); chronic daily administration led to significant reductions in both diffuse and neuritic plaques. These effects were observed in the absence of Notch-related changes (e.g., intestinal proliferation of goblet cells), which are commonly associated with repeated exposure to functional gamma-secretase inhibitors (GSIs).


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Anticuerpos/farmacología , Butiratos/farmacología , Cadherinas/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Transferencia Resonante de Energía de Fluorescencia/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrocarburos Halogenados/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Ratas , Receptores Notch/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Transfección/métodos
11.
J Clin Invest ; 119(12): 3692-702, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19907078

RESUMEN

Ectopic cell cycle events (CCEs) mark vulnerable neuronal populations in human Alzheimer disease (AD) and are observed early in disease progression. In transgenic mouse models of AD, CCEs are found before the onset of beta-amyloid peptide (Abeta) deposition to form senile plaques, a hallmark of AD. Here, we have demonstrated that alterations in brain microglia occur coincidently with the appearance of CCEs in the R1.40 transgenic mouse model of AD. Furthermore, promotion of inflammation with LPS at young ages in R1.40 mice induced the early appearance of neuronal CCEs, whereas treatment with 2 different nonsteroidal antiinflammatory drugs (NSAIDs) blocked neuronal CCEs and alterations in brain microglia without altering amyloid precursor protein (APP) processing and steady-state Abeta levels. In addition, NSAID treatment of older R1.40 animals prevented new neuronal CCEs, although it failed to reverse existing ones. Retrospective human epidemiological studies have identified long-term use of NSAIDs as protective against AD. Prospective clinical trials, however, have failed to demonstrate a similar benefit. Our use of CCEs as an outcome measure offers fresh insight into this discrepancy and provides important information for future clinical trials, as it suggests that NSAID use in human AD may need to be initiated as early as possible to prevent disease progression.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ciclo Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ibuprofeno/farmacología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Mutación , Naproxeno/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
J Biol Chem ; 284(6): 3793-803, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19074428

RESUMEN

Alzheimer disease beta-amyloid (Abeta) peptides are generated via sequential proteolysis of amyloid precursor protein (APP) by BACE1 and gamma-secretase. A subset of BACE1 localizes to cholesterol-rich membrane microdomains, termed lipid rafts. BACE1 processing in raft microdomains of cultured cells and neurons was characterized in previous studies by disrupting the integrity of lipid rafts by cholesterol depletion. These studies found either inhibition or elevation of Abeta production depending on the extent of cholesterol depletion, generating controversy. The intricate interplay between cholesterol levels, APP trafficking, and BACE1 processing is not clearly understood because cholesterol depletion has pleiotropic effects on Golgi morphology, vesicular trafficking, and membrane bulk fluidity. In this study, we used an alternate strategy to explore the function of BACE1 in membrane microdomains without altering the cellular cholesterol level. We demonstrate that BACE1 undergoes S-palmitoylation at four Cys residues at the junction of transmembrane and cytosolic domains, and Ala substitution at these four residues is sufficient to displace BACE1 from lipid rafts. Analysis of wild type and mutant BACE1 expressed in BACE1 null fibroblasts and neuroblastoma cells revealed that S-palmitoylation neither contributes to protein stability nor subcellular localization of BACE1. Surprisingly, non-raft localization of palmitoylation-deficient BACE1 did not have discernible influence on BACE1 processing of APP or secretion of Abeta. These results indicate that post-translational S-palmitoylation of BACE1 is not required for APP processing, and that BACE1 can efficiently cleave APP in both raft and non-raft microdomains.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Microdominios de Membrana/metabolismo , Ácidos Palmíticos/metabolismo , Procesamiento Proteico-Postraduccional , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Colesterol/genética , Colesterol/metabolismo , Estabilidad de Enzimas/genética , Estabilidad de Enzimas/fisiología , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Humanos , Microdominios de Membrana/genética , Ratones , Ratones Noqueados , Transporte de Proteínas/genética
13.
J Biol Chem ; 284(3): 1373-84, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19028695

RESUMEN

Proteolytic processing of amyloid precursor protein (APP) by beta- and gamma-secretases generates beta-amyloid (Abeta) peptides, which accumulate in the brains of individuals affected by Alzheimer disease. Detergent-resistant membrane microdomains (DRM) rich in cholesterol and sphingolipid, termed lipid rafts, have been implicated in Abeta production. Previously, we and others reported that the four integral subunits of the gamma-secretase associate with DRM. In this study we investigated the mechanisms underlying DRM association of gamma-secretase subunits. We report that in cultured cells and in brain the gamma-secretase subunits nicastrin and APH-1 undergo S-palmitoylation, the post-translational covalent attachment of the long chain fatty acid palmitate common in lipid raft-associated proteins. By mutagenesis we show that nicastrin is S-palmitoylated at Cys(689), and APH-1 is S-palmitoylated at Cys(182) and Cys(245). S-Palmitoylation-defective nicastrin and APH-1 form stable gamma-secretase complexes when expressed in knock-out fibroblasts lacking wild type subunits, suggesting that S-palmitoylation is not essential for gamma-secretase assembly. Nevertheless, fractionation studies show that S-palmitoylation contributes to DRM association of nicastrin and APH-1. Moreover, pulse-chase analyses reveal that S-palmitoylation is important for nascent polypeptide stability of both proteins. Co-expression of S-palmitoylation-deficient nicastrin and APH-1 in cultured cells neither affects Abeta40, Abeta42, and AICD production, nor intramembrane processing of Notch and N-cadherin. Our findings suggest that S-palmitoylation plays a role in stability and raft localization of nicastrin and APH-1, but does not directly modulate gamma-secretase processing of APP and other substrates.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Lipoilación/fisiología , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/enzimología , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Endopeptidasas , Estabilidad de Enzimas/fisiología , Humanos , Glicoproteínas de Membrana/genética , Lípidos de la Membrana/genética , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/genética , Proteínas de la Membrana/genética , Ratones , Péptido Hidrolasas , Receptores Notch/genética , Receptores Notch/metabolismo
14.
Mol Neurodegener ; 2: 4, 2007 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-17288597

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is characterized by cerebral deposition of beta-amyloid (Abeta) peptides. Abeta is released from ectodomain cleaved amyloid precursor protein (APP) via intramembranous proteolysis by gamma-secretase, a complex consisting of presenilin and a few other proteins. p23/TMP21, a member of the p24 family type I transmembrane proteins, was recently identified as a presenilin complex component capable of modulating gamma-secretase cleavage. The p24 family proteins form oligomeric complexes and regulate vesicular trafficking in the early secretory pathway, but their role in APP trafficking has not been investigated. RESULTS: Here, we report that siRNA-mediated depletion of p23 in N2a neuroblastoma and HeLa cells produces concomitant knockdown of additional p24 family proteins and increases secretion of sAPP. Furthermore, intact cell and cell-free Abeta production increases following p23 knockdown, similar to data reported earlier using HEK293 cells. However, we find that p23 is not present in mature gamma-secretase complexes isolated using an active-site gamma-secretase inhibitor. Depletion of p23 and expression of a familial AD-linked PS1 mutant have additive effects on Abeta42 production. Knockdown of p23 expression confers biosynthetic stability to nascent APP, allowing its efficient maturation and surface accumulation. Moreover, immunoisolation analyses show decrease in co-residence of APP and the APP adaptor Mint3. Thus, multiple lines of evidence indicate that p23 function influences APP trafficking and sAPP release independent of its reported role in gamma-secretase modulation. CONCLUSION: These data assign significance to p24 family proteins in regulating APP trafficking in the continuum of bidirectional transport between the ER and Golgi, and ascribe new relevance to the regulation of early trafficking in AD pathogenesis.

15.
J Biol Chem ; 281(43): 32240-53, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16945923

RESUMEN

Ubiquilin 1 (UBQLN1) is a ubiquitin-like protein, which has been shown to play a central role in regulating the proteasomal degradation of various proteins, including the presenilins. We recently reported that DNA variants in UBQLN1 increase the risk for Alzheimer disease, by influencing expression of this gene in brain. Here we present the first assessment of the effects of UBQLN1 on the metabolism of the amyloid precursor protein (APP). For this purpose, we employed RNA interference to down-regulate UBQLN1 in a variety of neuronal and non-neuronal cell lines. We demonstrate that down-regulation of UBQLN1 accelerates the maturation and intracellular trafficking of APP, while not interfering with alpha-, beta-, or gamma-secretase levels or activity. UBQLN1 knockdown increased the ratio of APP mature/immature, increased levels of full-length APP on the cell surface, and enhanced the secretion of sAPP (alpha- and beta-forms). Moreover, UBQLN1 knockdown increased levels of secreted Abeta40 and Abeta42. Finally, employing a fluorescence resonance energy transfer-based assay, we show that UBQLN1 and APP come into close proximity in intact cells, independently of the presence of the presenilins. Collectively, our findings suggest that UBQLN1 may normally serve as a cytoplasmic "gatekeeper" that may control APP trafficking from intracellular compartments to the cell surface. These findings suggest that changes in UBQLN1 steady-state levels affect APP trafficking and processing, thereby influencing the generation of Abeta.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Regulación hacia Abajo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Relacionadas con la Autofagia , Biotinilación , Proteínas Portadoras/química , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Células Cultivadas , Exones , Fibroblastos/citología , Fibroblastos/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Glioma/metabolismo , Glioma/patología , Humanos , Inmunohistoquímica , Ratones , Neuronas/citología , Neuronas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transfección
16.
Biochem Pharmacol ; 69(4): 689-98, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15670587

RESUMEN

A primary pathological feature of Alzheimer's disease is beta-amyloid (Abeta)-containing plaques in brain and cerebral vasculature. Reductions in the formation of Abeta peptides by gamma-secretase inhibitors may be a viable therapy for reducing Abeta in Alzheimer's disease. Here we report on the effects of two orally active gamma-secretase inhibitors. BMS-289948 (4-chloro-N-(2,5-difluorophenyl)-N-((1R)-{4-fluoro-2-[3-(1H-imidazol-1-yl)propyl]phenyl}ethyl)benzenesulfonamide hydrochloride) and BMS-299897 (4-[2-((1R)-1-{[(4-chlorophenyl)sulfonyl]-2,5-difluoroanilino}ethyl)-5-fluorophenyl]butanoic acid) markedly reduced both brain and plasma Abeta(1-40) in APP-YAC mice with ED(50) values of 86 and 22 mg/kg per os (po), respectively, for BMS-289948, and 30 and 16 mg/kg po, respectively, for BMS-299897. Both compounds also dose-dependently increased brain concentrations of APP carboxy-terminal fragments, consistent with inhibition of gamma-secretase. BMS-289948 and BMS-299897 (100 mg/kg po) reduced brain and plasma Abeta(1-40) rapidly (within 20min) and maximally within 3 h. BMS-299897 also dose-dependently reduced cortical, cerebrospinal fluid (CSF), and plasma Abeta in guinea pigs with ED(50) values of 30 mg/kg intraperitoneally, without affecting CSF levels of alpha-sAPP. The reductions in cortical Abeta correlated significantly with the reductions in both plasma (r(2) = 0.77) and CSF (r(2) = 0.61) Abeta. The decreases in Abeta were apparent at 3 and 6 h post-administration of BMS-299897, but not at 12h. These results demonstrate that BMS-289948 and BMS-299897 are orally bioavailable, functional gamma-secretase inhibitors with the ability to markedly reduce Abeta peptide concentrations in APP-YAC transgenic mice and in guinea pigs. These compounds may be useful pharmacologically for examining the effects of reductions in beta-amyloid peptides in both animal models and in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/análisis , Butiratos/farmacología , Endopeptidasas/efectos de los fármacos , Hidrocarburos Halogenados/farmacología , Imidazoles/farmacología , Inhibidores de Proteasas/farmacología , Sulfonamidas/farmacología , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/sangre , Animales , Ácido Aspártico Endopeptidasas , Química Encefálica/efectos de los fármacos , Femenino , Cobayas , Humanos , Masculino , Ratones , Fragmentos de Péptidos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...