Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Biol Chem ; 299(7): 104846, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37211092

RESUMEN

Apolipoprotein E (apoE) interaction with amyloid ß-protein precursor (APP) has garnered attention as the therapeutic target for Alzheimer's disease (AD). Having discovered the apoE antagonist (6KApoEp) that blocks apoE binding to N-terminal APP, we tested the therapeutic potential of 6KApoEp on AD-relevant phenotypes in amyloid ß-protein precursor/presenilin 1 (APP/PS1) mice that express each human apoE isoform of apoE2, apoE3, or apoE4 (designated APP/PS1/E2, APP/PS1/E3, or APP/PS1/E4 mice). At 12 months of age, we intraperitoneally administered 6KApoEp (250 µg/kg) or vehicle once daily for 3 months. At 15 months of age, blockage of apoE and N-terminal APP interaction by 6KApoEp treatment improved cognitive impairment in most tests of learning and memory, including novel object recognition and maze tasks in APP/PS1/E2, APP/PS1/E3, and APP/PS1/E4 mice versus each vehicle-treated mouse line and did not alter behavior in nontransgenic littermates. Moreover, 6KApoEp therapy ameliorated brain parenchymal and cerebral vascular ß-amyloid deposits and decreased abundance of amyloid ß-protein (Aß) in APP/PS1/E2, APP/PS1/E3, and APP/PS1/E4 mice versus each vehicle-treated mouse group. Notably, the highest effect in Aß-lowering by 6KApoEp treatment was observed in APP/PS1/E4 mice versus APP/PS1/E2 or APP/PS1/E3 mice. These effects occured through shifting toward lessened amyloidogenic APP processing due to decreasing APP abundance at the plasma membrane, reducing APP transcription, and inhibiting p44/42 mitogen-activated protein kinase phosphorylation. Our findings provide the preclinical evidence that 6KApoEp therapy aimed at targeting apoE and N-terminal APP interaction is a promising strategy and may be suitable for patients with AD carrying the apoE4 isoform.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Animales , Humanos , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/genética , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
2.
Int J Biol Sci ; 18(14): 5230-5240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147474

RESUMEN

Checkpoint immunotherapy is capable of unleashing T cells for controlling tumor, whereas it is destroyed by immunosuppressive myeloid cell. Apoprotein E (APOE) refers to a ligand in terms of the members of low-density lipoprotein (LDL) receptor family for mediating Apoprotein B-involving atherogenic lipoprotein clearance. Besides, tumor-infiltration macrophage can express APOE. The present study reported Apoe-/- mice to exhibit higher resistance toward the development of three types of carcinomas as compared with mice with wild type and to have greater responses to αPD-1 (anti-PD-1) immunotherapy. Moreover, treatment by exploiting APOE inhibitor (COG 133TFA, αAPOE) was capable of curbing tumor development and fostering regression if in combination of αPD-1. According to single-cell RNA sequencing (scRNA-seq), Apoe deletion was correlated with the decline of C1QC+ and CCR2+ macrophage within tumor infiltration, and mass spectrometry results noticeably showed down-regulated the number of M2 macrophages as well. Furthermore, APOE expression in cancer patients resistant to αPD-1 treatment significantly exceeded that in the sensitive group. For this reason, APOE is likely to be targeted for modifying tumor macrophage infiltrate and augmenting checkpoint immunotherapy.


Asunto(s)
Apolipoproteínas E , Apoproteínas , Inhibidores de Puntos de Control Inmunológico , Neoplasias , Animales , Apolipoproteínas E/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ligandos , Lipoproteínas LDL , Ratones , Ratones Noqueados para ApoE , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
4.
Neurobiol Dis ; 138: 104784, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32027932

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder that is growing in prevalence globally. It is the only major cause of death without any effective pharmacological means to treat or slow progression. Inheritance of the ε4 allele of the Apolipoprotein (APO) E gene is the strongest genetic risk factor for late-onset AD. The interaction between APOE and amyloid ß (Aß) plays a key role in AD pathogenesis. The APOE-Aß interaction regulates Aß aggregation and clearance and therefore directly influences the development of amyloid plaques, congophilic amyloid angiopathy and subsequent tau related pathology. Relatively few AD therapeutic approaches have directly targeted the APOE-Aß interaction thus far. Here we review the critical role of APOE in the pathogenesis of AD and some of the most promising therapeutic approaches that focus on the APOE-Aß interaction.


Asunto(s)
Apolipoproteínas E/metabolismo , Placa Amiloide/metabolismo , Alelos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E4/antagonistas & inhibidores , Apolipoproteína E4/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Encéfalo/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Ratones
5.
Cell ; 179(2): 312-339, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564456

RESUMEN

Alzheimer disease (AD) is a heterogeneous disease with a complex pathobiology. The presence of extracellular ß-amyloid deposition as neuritic plaques and intracellular accumulation of hyperphosphorylated tau as neurofibrillary tangles remains the primary neuropathologic criteria for AD diagnosis. However, a number of recent fundamental discoveries highlight important pathological roles for other critical cellular and molecular processes. Despite this, no disease-modifying treatment currently exists, and numerous phase 3 clinical trials have failed to demonstrate benefits. Here, we review recent advances in our understanding of AD pathobiology and discuss current treatment strategies, highlighting recent clinical trials and opportunities for developing future disease-modifying therapies.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Placa Amiloide/metabolismo , Proteínas tau , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Vacunas contra el Alzheimer/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Ensayos Clínicos como Asunto , Humanos , Ratones , Proteínas tau/antagonistas & inhibidores , Proteínas tau/metabolismo
6.
JCI Insight ; 4(18)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31534056

RESUMEN

Age is a well-established risk factor for impaired bone fracture healing. Here, we identify a role for apolipoprotein E (ApoE) in age-associated impairment of bone fracture healing and osteoblast differentiation, and we investigate the mechanism by which ApoE alters these processes. We identified that, in both humans and mice, circulating ApoE levels increase with age. We assessed bone healing in WT and ApoE-/- mice after performing tibial fracture surgery: bone deposition was higher within fracture calluses from ApoE-/- mice. In vitro recombinant ApoE (rApoE) treatment of differentiating osteoblasts decreased cellular differentiation and matrix mineralization. Moreover, this rApoE treatment decreased osteoblast glycolytic activity while increasing lipid uptake and fatty acid oxidation. Using parabiosis models, we determined that circulating ApoE plays a strong inhibitory role in bone repair. Using an adeno-associated virus-based siRNA system, we decreased circulating ApoE levels in 24-month-old mice and demonstrated that, as a result, fracture calluses from these aged mice displayed enhanced bone deposition and mechanical strength. Our results demonstrate that circulating ApoE as an aging factor inhibits bone fracture healing by altering osteoblast metabolism, thereby identifying ApoE as a new therapeutic target for improving bone repair in the elderly.


Asunto(s)
Envejecimiento/sangre , Apolipoproteínas E/sangre , Apolipoproteínas E/genética , Curación de Fractura/fisiología , Osteoblastos/fisiología , Fracturas de la Tibia/fisiopatología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Animales , Apolipoproteínas E/antagonistas & inhibidores , Callo Óseo/diagnóstico por imagen , Callo Óseo/efectos de los fármacos , Callo Óseo/fisiopatología , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Estudios de Cohortes , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Curación de Fractura/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados para ApoE , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , Proteínas Recombinantes/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Fracturas de la Tibia/diagnóstico por imagen , Fracturas de la Tibia/cirugía , Microtomografía por Rayos X
7.
PLoS Pathog ; 15(8): e1007874, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31393946

RESUMEN

Hepatitis B virus (HBV) is a common cause of liver diseases, including chronic hepatitis, steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). HBV chronically infects about 240 million people worldwide, posing a major global health problem. The current standard antiviral therapy effectively inhibits HBV replication but does not eliminate the virus unlike direct-acting antivirals (DAA) for curing hepatitis C. Our previous studies have demonstrated that human apolipoprotein E (apoE) plays important roles in hepatitis C virus infection and morphogenesis. In the present study, we have found that apoE is also associated with HBV and is required for efficient HBV infection. An apoE-specific monoclonal antibody was able to capture HBV similar to anti-HBs. More importantly, apoE monoclonal antibody could effectively block HBV infection, resulting in a greater than 90% reduction of HBV infectivity. Likewise, silencing of apoE expression or knockout of apoE gene by CRISPR/Cas9 resulted in a greater than 90% reduction of HBV infection and more than 80% decrease of HBV production, which could be fully restored by ectopic apoE expression. However, apoE silencing or knockout did not significantly affect HBV DNA replication or the production of nonenveloped (naked) nucleocapsids. These findings demonstrate that human apoE promotes HBV infection and production. We speculate that apoE may also play a role in persistent HBV infection by evading host immune response similar to its role in the HCV life cycle and pathogenesis. Inhibitors interfering with apoE biogenesis, secretion, and/or binding to receptors may serve as antivirals for elimination of chronic HBV infection.


Asunto(s)
Apolipoproteínas E/metabolismo , Carcinoma Hepatocelular/virología , Anticuerpos contra la Hepatitis B/inmunología , Virus de la Hepatitis B/patogenicidad , Hepatitis B/virología , Neoplasias Hepáticas/virología , Replicación Viral , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Células Hep G2 , Hepatitis B/complicaciones , Virus de la Hepatitis B/inmunología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , ARN Interferente Pequeño/genética
8.
Biol Psychiatry ; 86(3): 208-220, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31208706

RESUMEN

BACKGROUND: The ɛ4 isoform of apolipoprotein E (apoE4) is a major genetic risk factor for the development of sporadic Alzheimer's disease (AD), and its modification has been an intense focus for treatment of AD during recent years. METHODS: We investigated the binding of apoE, a peptide corresponding to its low-density lipoprotein receptor binding domain (amino acids 133-152; ApoEp), and modified ApoEp to amyloid precursor protein (APP) and their effects on amyloid-ß (Aß) production in cultured cells. Having discovered a peptide (6KApoEp) that blocks the interaction of apoE with N-terminal APP, we investigated the effects of this peptide and ApoEp on AD-like pathology and behavioral impairment in 3XTg-AD and 5XFAD transgenic mice. RESULTS: ApoE and ApoEp, but not truncated apoE lacking the low-density lipoprotein receptor binding domain, physically interacted with N-terminal APP and thereby mediated Aß production. Interestingly, the addition of 6 lysine residues to the N-terminus of ApoEp (6KApoEp) directly inhibited apoE binding to N-terminal APP and markedly limited apoE- and ApoEp-mediated Aß generation, presumably through decreasing APP cellular membrane trafficking and p44/42 mitogen-activated protein kinase phosphorylation. Moreover, while promoting apoE interaction with APP by ApoEp exacerbated Aß and tau brain pathologies in 3XTg-AD mice, disrupting this interaction by 6KApoEp ameliorated cerebral Aß and tau pathologies, neuronal apoptosis, synaptic loss, and hippocampal-dependent learning and memory impairment in 5XFAD mice without altering cholesterol, low-density lipoprotein receptor, and apoE expression levels. CONCLUSIONS: These data suggest that disrupting apoE interaction with N-terminal APP may be a novel disease-modifying therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Encéfalo/patología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Apolipoproteínas E/genética , Células CHO , Cognición/efectos de los fármacos , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos
9.
Comput Biol Chem ; 79: 83-90, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30743160

RESUMEN

AIM: An integrated protocol of virtual screening involving molecular docking, pharmacophore probing, and simulations was established to identify small novel molecules targeting crucial residues involved in the variant apoE ε4 to mimic its behavior as apoE2 thereby eliminating the amyloid plaque accumulation and facilitating its clearance. MATERIALS AND METHODS: An excellent ligand-based and structure-based approach was made to identify common pharmacophoric features involving structure-based docking with respect to apoE ε4 leading to the development of apoE ε4 inhibitors possessing new scaffolds. An effort was made to design multiple-substituted triazine derivatives series bearing a novel scaffold. A structure-based pharmacophore mapping was developed to explore the binding sites of apoE ε4 which was taken into consideration. Subsequently, virtual screening, ADMET, DFT searches were at work to narrow down the proposed hits to be forwarded as a potential drug likes candidates. Further, the binding patterns of the best-proposed hits were studied and were forwarded for molecular dynamic simulations of 10 ns for its structural optimization. RESULTS: Selectivity profile for the most promising candidates was studied, revealing significantly C13 and C15 to be the most potent compounds. The proposed hits can be forwarded for further study against apoE ε4 involved in neurological disorder Alzheimer's.


Asunto(s)
Apolipoproteínas E/antagonistas & inhibidores , Teoría Funcional de la Densidad , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Simulación de Dinámica Molecular , Bibliotecas de Moléculas Pequeñas/farmacología , Apolipoproteínas E/genética , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad Cuantitativa , Bibliotecas de Moléculas Pequeñas/química
11.
Neuron ; 96(5): 1013-1023.e4, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29216448

RESUMEN

The apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset Alzheimer disease. Previous studies suggest that reduction of apoE levels through genetic manipulation can reduce Aß pathology. However, it is not clear how reduction of apoE levels after birth would affect amyloid deposition. We utilize an antisense oligonucleotide (ASO) to reduce apoE expression in the brains of APP/PS1-21 mice homozygous for the APOE-ε4 or APOE-ε3 allele. ASO treatment starting after birth led to a significant decrease in Aß pathology when assessed at 4 months. Interestingly, ASO treatment starting at the onset of amyloid deposition led to an increase in Aß plaque size and a reduction in plaque-associated neuritic dystrophy with no change in overall plaque load. These results suggest that lowering apoE levels prior to plaque deposition can strongly affect the initiation of Aß pathology while lowering apoE after Aß seeding modulates plaque size and toxicity.


Asunto(s)
Péptidos beta-Amiloides , Amiloidosis/tratamiento farmacológico , Apolipoproteínas E/antagonistas & inhibidores , Oligonucleótidos Antisentido/uso terapéutico , Envejecimiento/fisiología , Alelos , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/patología , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Placa Amiloide/patología , Placa Amiloide/prevención & control
12.
Neuropharmacology ; 108: 179-92, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27114256

RESUMEN

Apolipoprotein E (apoE) is a major cholesterol carrier that regulates lipid homeostasis by mediating lipid transport from one tissue or cell type to another. In the central neural system (CNS), apoE is mainly produced by astrocytes, and transports cholesterol to neurons via apoE receptors, which are members of the low-density lipoprotein receptor family. The APOEε4 gene is a strong genetic risk factor for late-onset sporadic Alzheimer's disease (AD), likely through its strong effect on the accumulation of amyloid-ß (Aß) peptide. ApoE protein levels in cerebrospinal fluid (CSF) and plasma are reduced in APOEε4 carriers and in patients with AD. Furthermore, altered cholesterol levels are also associated with the risk of AD. Aß accumulation, oligomerization and deposition in the brain are central to the pathogenesis of AD. Mounting evidence demonstrates that apoE and apoE receptors play important roles in these processes. Astrocyte-derived apoE is pivotal for cerebral cholesterol metabolism and clearance of Aß. Thus, we hypothesized that increased apoE in the brain may be an effective therapeutic strategy for AD. We report here that quercetin can significantly increase apoE levels by inhibiting apoE degradation in immortalized astrocytes. Importantly, we show that oral administration of quercetin significantly increased brain apoE and reduced insoluble Aß levels in the cortex of 5xFAD amyloid model mice. Our results demonstrate that quercetin increases apoE levels through a novel mechanism and can be explored as a novel class of drug for AD therapy.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antioxidantes/administración & dosificación , Apolipoproteínas E/metabolismo , Corteza Cerebral/metabolismo , Placa Amiloide/metabolismo , Quercetina/administración & dosificación , Administración Oral , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Apolipoproteínas E/antagonistas & inhibidores , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Línea Celular Transformada , Corteza Cerebral/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/tratamiento farmacológico , Estabilidad Proteica/efectos de los fármacos
13.
J Virol ; 89(19): 9962-73, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26202245

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) infects hepatocytes through two different routes: (i) cell-free particle diffusion followed by engagement with specific cellular receptors and (ii) cell-to-cell direct transmission mediated by mechanisms not well defined yet. HCV exits host cells in association with very-low-density lipoprotein (VLDL) components. VLDL particles contain apolipoproteins B (ApoB) and E (ApoE), which are required for viral assembly and/or infectivity. Based on these precedents, we decided to study whether these VLDL components participate in HCV cell-to-cell transmission in vitro. We observed that cell-to-cell viral spread was compromised after ApoE interference in donor but not in acceptor cells. In contrast, ApoB knockdown in either donor or acceptor cells did not impair cell-to-cell viral transmission. Interestingly, ApoB participated in the assembly of cell-free infective virions, suggesting a differential regulation of cell-to-cell and cell-free HCV infection. This study identifies host-specific factors involved in these distinct routes of infection that may unveil new therapeutic targets and advance our understanding of HCV pathogenesis. IMPORTANCE: This work demonstrates that cell-to-cell transmission of HCV depends on ApoE but not ApoB. The data also indicate that ApoB is required for the assembly of cell-free infective particles, strongly suggesting the existence of mechanisms involving VLDL components that differentially regulate cell-free and cell-to-cell HCV transmission. These data clarify some of the questions regarding the role of VLDL in HCV pathogenesis and the transmission of the virus cell to cell as a possible mechanism of immune evasion and open the door to therapeutic intervention.


Asunto(s)
Apolipoproteínas B/metabolismo , Apolipoproteínas E/metabolismo , Hepacivirus/patogenicidad , Hepatitis C/transmisión , Hepatocitos/metabolismo , Hepatocitos/virología , Apolipoproteínas B/antagonistas & inhibidores , Apolipoproteínas B/genética , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/genética , Línea Celular , Sistema Libre de Células , Técnicas de Silenciamiento del Gen , Hepacivirus/genética , Hepacivirus/fisiología , Hepatitis C/metabolismo , Hepatitis C/virología , Interacciones Huésped-Patógeno/fisiología , Humanos , Lipoproteínas VLDL/metabolismo , Modelos Biológicos , Ensamble de Virus/fisiología
14.
Nat Med ; 21(6): 628-37, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25985364

RESUMEN

Previous studies investigating the role of smooth muscle cells (SMCs) and macrophages in the pathogenesis of atherosclerosis have provided controversial results owing to the use of unreliable methods for clearly identifying each of these cell types. Here, using Myh11-CreER(T2) ROSA floxed STOP eYFP Apoe(-/-) mice to perform SMC lineage tracing, we find that traditional methods for detecting SMCs based on immunostaining for SMC markers fail to detect >80% of SMC-derived cells within advanced atherosclerotic lesions. These unidentified SMC-derived cells exhibit phenotypes of other cell lineages, including macrophages and mesenchymal stem cells (MSCs). SMC-specific conditional knockout of Krüppel-like factor 4 (Klf4) resulted in reduced numbers of SMC-derived MSC- and macrophage-like cells, a marked reduction in lesion size, and increases in multiple indices of plaque stability, including an increase in fibrous cap thickness as compared to wild-type controls. On the basis of in vivo KLF4 chromatin immunoprecipitation-sequencing (ChIP-seq) analyses and studies of cholesterol-treated cultured SMCs, we identified >800 KLF4 target genes, including many that regulate pro-inflammatory responses of SMCs. Our findings indicate that the contribution of SMCs to atherosclerotic plaques has been greatly underestimated, and that KLF4-dependent transitions in SMC phenotype are critical in lesion pathogenesis.


Asunto(s)
Aterosclerosis/genética , Factores de Transcripción de Tipo Kruppel/genética , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/genética , Animales , Apolipoproteínas E/antagonistas & inhibidores , Aterosclerosis/patología , Diferenciación Celular/genética , Linaje de la Célula , Rastreo Celular , Humanos , Factor 4 Similar a Kruppel , Macrófagos/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Placa Aterosclerótica/patología , Regiones Promotoras Genéticas
15.
PLoS Pathog ; 11(4): e1004859, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25923687

RESUMEN

It is well established that persistent viral infection may impair cellular function of specialized cells without overt damage. This concept, when applied to neurotropic viruses, may help to understand certain neurologic and neuropsychiatric diseases. Borna disease virus (BDV) is an excellent example of a persistent virus that targets the brain, impairs neural functions without cell lysis, and ultimately results in neurobehavioral disturbances. Recently, we have shown that BDV infects human neural progenitor cells (hNPCs) and impairs neurogenesis, revealing a new mechanism by which BDV may interfere with brain function. Here, we sought to identify the viral proteins and molecular pathways that are involved. Using lentiviral vectors for expression of the bdv-p and bdv-x viral genes, we demonstrate that the phosphoprotein P, but not the X protein, diminishes human neurogenesis and, more particularly, GABAergic neurogenesis. We further reveal a decrease in pro-neuronal factors known to be involved in neuronal differentiation (ApoE, Noggin, TH and Scg10/Stathmin2), demonstrating that cellular dysfunction is associated with impairment of specific components of the molecular program that controls neurogenesis. Our findings thus provide the first evidence that a viral protein impairs GABAergic human neurogenesis, a process that is dysregulated in several neuropsychiatric disorders. They improve our understanding of the mechanisms by which a persistent virus may interfere with brain development and function in the adult.


Asunto(s)
Virus de la Enfermedad de Borna/fisiología , Regulación hacia Abajo , Neuronas GABAérgicas/metabolismo , Interacciones Huésped-Patógeno , Neurogénesis , Fosfoproteínas/metabolismo , Proteínas Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Biomarcadores/química , Biomarcadores/metabolismo , Enfermedad de Borna/metabolismo , Enfermedad de Borna/patología , Enfermedad de Borna/virología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proliferación Celular , Células Cultivadas , Francia , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/patología , Neuronas GABAérgicas/virología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/patología , Células Madre Embrionarias Humanas/virología , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Estatmina , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/metabolismo , Proteínas Estructurales Virales/genética
16.
ACS Comb Sci ; 17(3): 196-201, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25602927

RESUMEN

A series of linear and cyclic peptoids, which were expected to possess better pharmacokinetic properties and biological activities for blocking the interaction between apolipoprotein E and amyloid-ß, were designed and synthesized as possible therapeutic agents. Peptoids were easily synthesized on solid-phase by the submonomer strategy and polar side chain-containing amines were effectively introduced under the modified reaction conditions. For the synthesis of cyclic peptoids, ß-alanine protected with the 2-phenylisopropyl group, which could be selectively removed by 2% TFA, was used as a primary amine to afford a complete peptoid unit. The macrolactamization between the carboxylic acid of ß-alanine moiety and terminal amine of peptoids was successfully performed in the presence of the PyAOP coupling agent on solid-phase in all the cases, providing various sizes of cyclic peptoids. In particular, some cyclic peptoids prepared in this study are the largest in size among cyclic peptoids reported to date. The synthetic strategy which was adopted in this study can also provide a robust platform for solid-phase construction of cyclic peptoid libraries. Currently, synthetic peptoids have been used to test interesting biological activities including the ApoE/Aß interaction inhibition, nontoxicity, the blood-brain barrier permeability, etc.


Asunto(s)
Peptoides/síntesis química , Peptoides/farmacología , Técnicas de Síntesis en Fase Sólida/métodos , Péptidos beta-Amiloides/antagonistas & inhibidores , Apolipoproteínas E/antagonistas & inhibidores , Estructura Molecular , Peptoides/química , Relación Estructura-Actividad
17.
J Neurochem ; 128(4): 577-91, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24117759

RESUMEN

Inheritance of the apolipoprotein E4 (apoE4) genotype has been identified as the major genetic risk factor for late-onset Alzheimer's disease (AD). Studies have shown that the binding between apoE and amyloid-ß (Aß) peptides occurs at residues 244-272 of apoE and residues 12-28 of Aß. ApoE4 has been implicated in promoting Aß deposition and impairing clearance of Aß. We hypothesized that blocking the apoE/Aß interaction would serve as an effective new approach to AD therapy. We have previously shown that treatment with Aß12-28P can reduce amyloid plaques in APP/PS1 transgenic (Tg) mice and vascular amyloid in TgSwDI mice with congophilic amyloid angiopathy. In the present study, we investigated whether the Aß12-28P elicits a therapeutic effect on tau-related pathology in addition to amyloid pathology using old triple transgenic AD mice (3xTg, with PS1M146V , APPSwe and tauP30IL transgenes) with established pathology from the ages of 21 to 26 months. We show that treatment with Aß12-28P substantially reduces tau pathology both immunohistochemically and biochemically, as well as reducing the amyloid burden and suppressing the activation of astrocytes and microglia. These affects correlate with a behavioral amelioration in the treated Tg mice.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Amiloidosis/tratamiento farmacológico , Apolipoproteínas E/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/farmacología , Amiloidosis/patología , Amiloidosis/psicología , Animales , Western Blotting , Química Encefálica/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Conducta Exploratoria/fisiología , Gliosis/patología , Inmunohistoquímica , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Actividad Motora/fisiología , Equilibrio Postural/efectos de los fármacos
18.
Nat Rev Neurol ; 9(2): 106-18, 2013 02.
Artículo en Inglés | MEDLINE | ID: mdl-23296339

RESUMEN

Apolipoprotein E (Apo-E) is a major cholesterol carrier that supports lipid transport and injury repair in the brain. APOE polymorphic alleles are the main genetic determinants of Alzheimer disease (AD) risk: individuals carrying the ε4 allele are at increased risk of AD compared with those carrying the more common ε3 allele, whereas the ε2 allele decreases risk. Presence of the APOE ε4 allele is also associated with increased risk of cerebral amyloid angiopathy and age-related cognitive decline during normal ageing. Apo-E-lipoproteins bind to several cell-surface receptors to deliver lipids, and also to hydrophobic amyloid-ß (Aß) peptide, which is thought to initiate toxic events that lead to synaptic dysfunction and neurodegeneration in AD. Apo-E isoforms differentially regulate Aß aggregation and clearance in the brain, and have distinct functions in regulating brain lipid transport, glucose metabolism, neuronal signalling, neuroinflammation, and mitochondrial function. In this Review, we describe current knowledge on Apo-E in the CNS, with a particular emphasis on the clinical and pathological features associated with carriers of different Apo-E isoforms. We also discuss Aß-dependent and Aß-independent mechanisms that link Apo-E4 status with AD risk, and consider how to design effective strategies for AD therapy by targeting Apo-E.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Enfermedad de Alzheimer/terapia , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Encéfalo/patología , Lesiones Encefálicas/genética , Lesiones Encefálicas/fisiopatología , Disfunción Cognitiva/patología , Disfunción Cognitiva/psicología , Demencia/genética , Demencia/fisiopatología , Espinas Dendríticas/fisiología , Humanos , Inmunoterapia , Inflamación/genética , Inflamación/patología , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Neurogénesis/genética , Neurogénesis/fisiología , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Valor Predictivo de las Pruebas , Riesgo , Enfermedades Vasculares/genética , Enfermedades Vasculares/fisiopatología , Proteínas tau/metabolismo
19.
J Neurosci ; 32(14): 4803-11, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22492035

RESUMEN

Apolipoprotein E4 (apoE4) plays a major role in the pathogenesis of Alzheimer's disease. Brain amyloid-ß (Aß) accumulation depends on age and apoE isoforms (apoE4 > apoE3) both in humans and in transgenic mouse models. Brain apoE levels are also isoform dependent, but in the opposite direction (apoE4 < apoE3). Thus, one prevailing hypothesis is to increase brain apoE expression to reduce Aß levels. To test this hypothesis, we generated mutant human amyloid precursor protein transgenic mice expressing one or two copies of the human APOE3 or APOE4 gene that was knocked in and flanked by LoxP sites. We report that reducing apoE3 or apoE4 expression by 50% in 6-month-old mice results in efficient Aß clearance and does not increase Aß accumulation. However, 12-month-old mice with one copy of the human APOE gene had significantly reduced Aß levels and plaque loads compared with mice with two copies, regardless of which human apoE isoform was expressed, suggesting a gene dose-dependent effect of apoE on Aß accumulation in aged mice. Additionally, 12-month-old mice expressing one or two copies of the human APOE4 gene had significantly higher levels of Aß accumulation and plaque loads than age-matched mice expressing one or two copies of the human APOE3 gene, suggesting an isoform-dependent effect of apoE on Aß accumulation in aged mice. Moreover, Cre-mediated APOE4 gene excision in hippocampal astrocytes significantly reduced insoluble Aß in adult mice. Thus, reducing, rather than increasing, apoE expression is an attractive approach to lowering brain Aß levels.


Asunto(s)
Envejecimiento/genética , Precursor de Proteína beta-Amiloide/genética , Apolipoproteína E3/deficiencia , Apolipoproteína E4/deficiencia , Apolipoproteínas E/deficiencia , Mutación/genética , Envejecimiento/fisiología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/fisiología , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...