Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
AAPS PharmSciTech ; 24(8): 223, 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37945928

RESUMEN

Rivastigmine hydrogen tartrate (RHT) is an acetylcholinesterase (AChE) inhibitor used in the management of Alzheimer's disease (AD). RHT is a BCS class-I drug that undergoes significant first-pass metabolism. Permeating a hydrophilic drug through the brain remains a major challenge in brain delivery. In this study, the RHT was incorporated inside the hydrophilic core of liposomes (LPS) and then coated with the ApoE3. ApoE3-coated RHT-loaded liposomes (ApoE3-RHT-LPS) were fabricated through the thin film hydration method using DSPE-PEG. The coating of LPS with ApoE3 enhances brain uptake and improves Aß clearance. The results obtained from the physicochemical characterization demonstrated that ApoE3-RHT-LPS shows a particle size of 128.6 ± 2.16 nm and a zeta potential of 16.6 ± 1.19. The % entrapment efficiency and % drug loading were found to be 75% and 17.84%, respectively. The data obtained from TEM and SEM studies revealed that the particle size of the LPS was less than 200 nm. An in vitro AChE assay was performed, and the results demonstrated the AChE inhibitory potential of ApoE3-RHT-LPS. Through the intravenous route, an in vivo pharmacokinetic study of formulation displayed improved brain uptake of RHT by ~ 1.3-fold than pure RHT due to ApoE3 coating. In vivo, biodistribution studies in vital organs suggested that the biodistribution of RHT to the liver was significantly reduced (p < 0.001), signifying an increase in the drug's half-life and blood circulation time. All research findings suggested that ApoE3 coating and LPS strategy are proven effective for improving the brain uptake of RHT designed for the management of AD.


Asunto(s)
Enfermedad de Alzheimer , Liposomas , Humanos , Rivastigmina , Liposomas/química , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Acetilcolinesterasa/metabolismo , Acetilcolinesterasa/farmacología , Acetilcolinesterasa/uso terapéutico , Distribución Tisular , Lipopolisacáridos , Encéfalo/metabolismo , Inhibidores de la Colinesterasa , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Tamaño de la Partícula
2.
Int J Obes (Lond) ; 47(3): 236-243, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36732416

RESUMEN

OBJECTIVES: Studies in mice have recently linked increased dietary choline consumption to increased incidence of obesity-related metabolic diseases, while several clinical trials have reported an anti-obesity effect of high dietary choline intake. Since the underlying mechanisms by which choline affects obesity are incompletely understood, the aim of the present study was to investigate the role of dietary choline supplementation in adiposity. METHODS: Female APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism and cardiometabolic diseases, were fed a Western-type diet supplemented with or without choline (1.2%, w/w) for up to 16 weeks. RESULTS: Dietary choline reduced body fat mass gain, prevented adipocyte enlargement, and attenuated adipose tissue inflammation. Besides, choline ameliorated liver steatosis and damage, associated with an upregulation of hepatic genes involved in fatty acid oxidation. Moreover, choline reduced plasma cholesterol, as explained by a reduction of plasma non-HDL cholesterol. Mechanistically, choline reduced hepatic VLDL-cholesterol secretion and enhanced the selective uptake of fatty acids from triglyceride-rich lipoprotein (TRL)-like particles by brown adipose tissue (BAT), consequently accelerating the clearance of the cholesterol-enriched TRL remnants by the liver. CONCLUSIONS: In APOE*3-Leiden.CETP mice, dietary choline reduces body fat by enhancing TRL-derived fatty acids by BAT, resulting in accelerated TRL turnover to improve hypercholesterolemia. These data provide a mechanistic basis for the observation in human intervention trials that high choline intake is linked with reduced body weight.


Asunto(s)
Tejido Adiposo Pardo , Colina , Ratones , Femenino , Humanos , Animales , Tejido Adiposo Pardo/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Colina/farmacología , Colina/metabolismo , Colesterol , Triglicéridos , Lipoproteínas/metabolismo , Lipoproteínas/farmacología , Hígado/metabolismo , Dieta , Tejido Adiposo/metabolismo , Obesidad/metabolismo , Ácidos Grasos/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo
3.
Neuropeptides ; 97: 102307, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36434832

RESUMEN

Apolipoprotein E (ApoE) is the main cholesterol carrier of the brain and the ε4 gene variant (APOE4) is the most prevalent genetic risk factor for Alzheimer's disease (AD), increasing risk up to 15-fold. Several studies indicate that APOE4 modulates critical factors for neuronal function, including brain-derived neurotrophic factor (BDNF) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). Both proteins show exercise-induced upregulation, which is presumed to mediate many of the beneficial effects of physical activity including improved cognition; however, there is variability in results between individuals potentially in-part due to genetic variations including APOE isoform. This study aimed to determine if the two most prevalent human APOE isoforms influence adaptive responses to exercise-training. Targeted replacement mice, homozygous for either APOE3 or APOE4 were randomized into exercised and sedentary groups. Baseline locomotor function and voluntary wheel-running behavior was reduced in APOE4 mice. Exercised groups were subjected to daily treadmill running for 8 weeks. ApoE protein in brain cortex was significantly increased by exercise in both genotypes. PGC-1α mRNA levels in brain cortex were significantly lower in APOE4 mice, and only tended to increase with exercise in both genotypes. Hippocampal BDNF protein were similar between genotypes and was not significantly modulated by treadmill running. Behavioral and biochemical variations between APOE3 and APOE4 mice likely contribute to the differential risk for neurological and vascular diseases and the exercise-induced increase in ApoE levels suggests an added feature of the potential efficacy of physical activity as a preventative and therapeutic strategy for neurogenerative processes in both genotypes.


Asunto(s)
Apolipoproteína E4 , Factor Neurotrófico Derivado del Encéfalo , Ratones , Femenino , Animales , Humanos , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E4/farmacología , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratones Transgénicos , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacología , Encéfalo/metabolismo
4.
Oxid Med Cell Longev ; 2022: 2520348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36425057

RESUMEN

Influenza virus infection is one of the strongest pathogenic factors for the development of acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS). However, the underlying cellular and molecular mechanisms have not been clarified. In this study, we aim to investigate whether melatonin modulates macrophage polarization, oxidative stress, and pyroptosis via activating Apolipoprotein E/low-density lipoprotein receptor (ApoE/LDLR) pathway in influenza A-induced ALI. Here, wild-type (WT) and ApoE-/- mice were instilled intratracheally with influenza A (H3N2) and injected intraperitoneally with melatonin for 7 consecutive days. In vitro, WT and ApoE-/- murine bone marrow-derived macrophages (BMDMs) were pretreated with melatonin before H3N2 stimulation. The results showed that melatonin administration significantly attenuated H3N2-induced pulmonary damage, leukocyte infiltration, and edema; decreased the expression of proinflammatory M1 markers; enhanced anti-inflammatory M2 markers; and switched the polarization of alveolar macrophages (AMs) from M1 to M2 phenotype. Additionally, melatonin inhibited reactive oxygen species- (ROS-) mediated pyroptosis shown by downregulation of malonaldehyde (MDA) and ROS levels as well as inhibition of the NLRP3/GSDMD pathway and lactate dehydrogenase (LDH) release. Strikingly, the ApoE/LDLR pathway was activated when melatonin was applied in H3N2-infected macrophages and mice. ApoE knockout mostly abrogated the protective impacts of melatonin on H3N2-induced ALI and its regulatory ability on macrophage polarization, oxidative stress, and pyroptosis. Furthermore, recombinant ApoE3 (re-ApoE3) inhibited H3N2-induced M1 polarization of BMDMs with upregulation of MT1 and MT2 expression, but re-ApoE2 and re-ApoE4 failed to do this. Melatonin combined with re-ApoE3 played more beneficial protective effects on modulating macrophage polarization, oxidative stress, and pyroptosis in H3N2-infected ApoE-/- BMDMs. Our study indicated that melatonin attenuated influenza A- (H3N2-) induced ALI by inhibiting the M1 polarization of pulmonary macrophages and ROS-mediated pyroptosis via activating the ApoE/LDLR pathway. This study suggested that melatonin-ApoE/LDLR axis may serve as a novel therapeutic strategy for influenza virus-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda , Melatonina , Infecciones por Orthomyxoviridae , Animales , Ratones , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/virología , Apolipoproteína E3/farmacología , Apolipoproteínas E/metabolismo , Subtipo H3N2 del Virus de la Influenza A , Macrófagos/metabolismo , Melatonina/uso terapéutico , Ratones Noqueados para ApoE , Piroptosis , Especies Reactivas de Oxígeno/metabolismo , Infecciones por Orthomyxoviridae/tratamiento farmacológico
5.
Neurobiol Aging ; 118: 13-24, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35843109

RESUMEN

Female APOE4 carriers are at greatest risk of Alzheimer's disease (AD). The potent estrogen 17ß-estradiol (E2) may mediate AD risk, as the onset of memory decline coincides with the menopausal transition. Whether APOE genotype mediates E2's effects on memory and neuronal morphology is poorly understood. We used the APOE+/+/5xFAD+/- (EFAD) mouse model to examine how APOE3 homozygote (E3FAD), APOE3/4 heterozygote (E3/4FAD), and APOE4 homozygote (E4FAD) genotypes modulate effects of E2 on object and spatial memory consolidation, dendritic spine density, and dorsal hippocampal estrogen receptor expression in 6-month-old ovariectomized EFAD mice. Dorsal hippocampal E2 infusion enhanced memory consolidation and increased CA1 apical spine density in E3FAD and E3/4FAD, but not E4FAD, mice. CA1 basal mushroom spines were also increased by E2 in E3FADs. E4FAD mice exhibited reduced CA1 and mPFC basal spine density, and increased dorsal hippocampal ERα protein, independent of E2. Overall, E2 benefitted hippocampal memory and structural plasticity in females bearing one or no APOE4 allele, whereas two APOE4 alleles impeded the memory-enhancing and spinogenic effects of E2.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Estradiol/metabolismo , Estradiol/farmacología , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Homocigoto , Ratones , Ratones Transgénicos
6.
Drug Metab Dispos ; 50(10): 1414-1428, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35878927

RESUMEN

Cadmium (Cd) exposure is associated with increased Alzheimer's disease (AD) risks. The human Apolipoprotein E (ApoE) gene encodes a lipid-transporting protein that is critical for brain functions. Compared with ApoE2 and E3, ApoE4 is associated with increased AD risk. Xenobiotic biotransformation-related genes have been implicated in the pathogenesis of AD. However, little is known about the effects of Cd, ApoE, and sex on drug-processing genes. We investigated the Cd-ApoE interaction on the transcriptomic changes in the brains and livers of ApoE3/ApoE4 transgenic mice. Cd disrupts the transcriptomes of transporter and drug-processing genes in brain and liver in a sex- and ApoE-genotype-specific manner. Proinflammation related genes were enriched in livers of Cd-exposed ApoE4 males, whereas circadian rhythm and lipid metabolism related genes were enriched in livers of Cd-exposed ApoE3 females. In brains, Cd up-regulated the arachidonic acid-metabolizing Cyp2j isoforms only in the brains of ApoE3 mice, whereas the dysregulation of cation transporters was male-specific. In livers, several direct target genes of the major xenobiotic-sensing nuclear receptor pregnane X receptor were uniquely upregulated in Cd-exposed ApoE4 males. There was a female-specific hepatic upregulation of the steroid hormone-metabolizing Cyp2 isoforms and the bile acid synthetic enzyme Cyp7a1 by Cd exposure. The dysregulated liver transporters were mostly involved in intermediary metabolism, with the most significant response observed in ApoE3 females. In conclusion, Cd dysregulated the brain and liver drug-processing genes in a sex- and ApoE-genotype specific manner, and this may serve as a contributing factor for the variance in the susceptibility to Cd neurotoxicity. SIGNIFICANCE STATEMENT: Xenobiotic biotransformation plays an important role in modulating the toxicity of environmental pollutants. The human ApoE4 allele is the strongest genetic risk factor for AD, and cadmium (Cd) is increasingly recognized as an environmental factor of AD. Very little is known regarding the interactions between Cd exposure, sex, and the genes involved in xenobiotic biotransformation in brain and liver. The present study has addressed this critical knowledge gap.


Asunto(s)
Enfermedad de Alzheimer , Contaminantes Ambientales , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Animales , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E2/farmacología , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E4/farmacología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacología , Ácido Araquidónico/metabolismo , Ácidos y Sales Biliares/metabolismo , Encéfalo/metabolismo , Cadmio/toxicidad , Contaminantes Ambientales/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Hormonas/metabolismo , Hormonas/farmacología , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Receptor X de Pregnano/metabolismo , Isoformas de Proteínas/metabolismo , Xenobióticos/metabolismo
7.
Metabolism ; 127: 154954, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34875308

RESUMEN

INTRODUCTION: Atherosclerotic Coronary Artery Disease (ASCAD) is the leading cause of mortality worldwide. Novel therapeutic approaches aiming to improve the atheroprotective functions of High Density Lipoprotein (HDL) include the use of reconstituted HDL forms containing human apolipoprotein A-I (rHDL-apoA-I). Given the strong atheroprotective properties of apolipoprotein E3 (apoE3), rHDL-apoE3 may represent an attractive yet largely unexplored therapeutic agent. OBJECTIVE: To evaluate the atheroprotective potential of rHDL-apoE3 starting with the unbiased assessment of global transcriptome effects and focusing on endothelial cell (EC) migration as a critical process in re-endothelialization and atherosclerosis prevention. The cellular, molecular and functional effects of rHDL-apoE3 on EC migration-associated pathways were assessed, as well as the potential translatability of these findings in vivo. METHODS: Human Aortic ECs (HAEC) were treated with rHDL-apoE3 and total RNA was analyzed by whole genome microarrays. Expression and phosphorylation changes of key EC migration-associated molecules were validated by qRT-PCR and Western blot analysis in primary HAEC, Human Coronary Artery ECs (HCAEC) and the human EA.hy926 EC line. The capacity of rHDL-apoE3 to stimulate EC migration was assessed by wound healing and transwell migration assays. The contribution of MEK1/2, PI3K and the transcription factor ID1 in rHDL-apoE3-induced EC migration and activation of EC migration-related effectors was assessed using specific inhibitors (PD98059: MEK1/2, LY294002: PI3K) and siRNA-mediated gene silencing, respectively. The capacity of rHDL-apoE3 to improve vascular permeability and hypercholesterolemia in vivo was tested in a mouse model of hypercholesterolemia (apoE KO mice) using Evans Blue assays and lipid/lipoprotein analysis in the serum, respectively. RESULTS: rHDL-apoE3 induced significant expression changes in 198 genes of HAEC mainly involved in re-endothelialization and atherosclerosis-associated functions. The most pronounced effect was observed for EC migration, with 42/198 genes being involved in the following EC migration-related pathways: 1) MEK/ERK, 2) PI3K/AKT/eNOS-MMP2/9, 3) RHO-GTPases, 4) integrin. rHDL-apoE3 induced changes in 24 representative transcripts of these pathways in HAEC, increasing the expression of their key proteins PIK3CG, EFNB2, ID1 and FLT1 in HCAEC and EA.hy926 cells. In addition, rHDL-apoE3 stimulated migration of HCAEC and EA.hy926 cells, and the migration was markedly attenuated in the presence of PD98059 or LY294002. rHDL-apoE3 also increased the phosphorylation of ERK1/2, AKT, eNOS and p38 MAPK in these cells, while PD98059 and LY294002 inhibited rHDL-apoE3-induced phosphorylation of ERK1/2, AKT and p38 MAPK, respectively. LY had no effect on rHDL-apoE3-mediated eNOS phosphorylation. ID1 siRNA markedly decreased EA.hy926 cell migration by inhibiting rHDL-apoE3-triggered ERK1/2 and AKT phosphorylation. Finally, administration of a single dose of rHDL-apoE3 in apoE KO mice markedly improved vascular permeability as demonstrated by the reduced concentration of Evans Blue dye in tissues such as the stomach, the tongue and the urinary bladder and ameliorated hypercholesterolemia. CONCLUSIONS: rHDL-apoE3 significantly enhanced EC migration in vitro, predominantly via overexpression of ID1 and subsequent activation of MEK1/2 and PI3K, and their downstream targets ERK1/2, AKT and p38 MAPK, respectively, and improved vascular permeability in vivo. These novel insights into the rHDL-apoE3 functions suggest a potential clinical use to promote re-endothelialization and retard development of atherosclerosis.


Asunto(s)
Apolipoproteína E3/farmacología , Células Endoteliales/efectos de los fármacos , Lipoproteínas HDL/farmacología , Animales , Apolipoproteína E3/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/fisiología , Humanos , Proteína 1 Inhibidora de la Diferenciación/antagonistas & inhibidores , Proteína 1 Inhibidora de la Diferenciación/efectos de los fármacos , Proteína 1 Inhibidora de la Diferenciación/genética , Lipoproteínas HDL/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Mol Neurodegener ; 16(1): 26, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863362

RESUMEN

BACKGROUND: Apolipoprotein E4 (APOE4) is associated with a greater response to neuroinflammation and the risk of developing late-onset Alzheimer's disease (AD), but the mechanisms for this association are not clear. The activation of calcium-dependent cytosolic phospholipase A2 (cPLA2) is involved in inflammatory signaling and is elevated within the plaques of AD brains. The relation between APOE4 genotype and cPLA2 activity is not known. METHODS: Mouse primary astrocytes, mouse and human brain samples differing by APOE genotypes were collected for measuring cPLA2 expression, phosphorylation, and activity in relation to measures of inflammation and oxidative stress. RESULTS: Greater cPLA2 phosphorylation, cPLA2 activity and leukotriene B4 (LTB4) levels were identified in ApoE4 compared to ApoE3 in primary astrocytes, brains of ApoE-targeted replacement (ApoE-TR) mice, and in human brain homogenates from the inferior frontal cortex of patients with AD carrying APOE3/E4 compared to APOE3/E3. Greater cPLA2 phosphorylation was also observed in human postmortem frontal cortical synaptosomes and primary astrocytes after treatment with recombinant ApoE4 ex vivo. In ApoE4 astrocytes, the greater levels of LTB4, reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) were reduced after cPLA2 inhibition. CONCLUSIONS: Our findings implicate greater activation of cPLA2 signaling system with APOE4, which could represent a potential drug target for mitigating the increased neuroinflammation with APOE4 and AD.


Asunto(s)
Apolipoproteína E4/metabolismo , Calcio/farmacología , Corteza Cerebral/enzimología , Sistema de Señalización de MAP Quinasas , Fosfolipasas A2 Citosólicas/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteína E4/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Corteza Cerebral/patología , Activación Enzimática/efectos de los fármacos , Heterocigoto , Humanos , Inflamasomas , Inflamación , Leucotrieno B4/biosíntesis , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo , Fragmentos de Péptidos/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno , Sinaptosomas/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
9.
Cell Rep ; 32(4): 107962, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726626

RESUMEN

Despite its clear impact on Alzheimer's disease (AD) risk, apolipoprotein (apo) E4's contributions to AD etiology remain poorly understood. Progress in answering this and other questions in AD research has been limited by an inability to model human-specific phenotypes in an in vivo environment. Here we transplant human induced pluripotent stem cell (hiPSC)-derived neurons carrying normal apoE3 or pathogenic apoE4 into human apoE3 or apoE4 knockin mouse hippocampi, enabling us to disentangle the effects of apoE4 produced in human neurons and in the brain environment. Using single-nucleus RNA sequencing (snRNA-seq), we identify key transcriptional changes specific to human neuron subtypes in response to endogenous or exogenous apoE4. We also find that Aß from transplanted human neurons forms plaque-like aggregates, with differences in localization and interaction with microglia depending on the transplant and host apoE genotype. These findings highlight the power of in vivo chimeric disease modeling for studying AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Apolipoproteína E4/metabolismo , Neuronas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Quimera/genética , Quimera/metabolismo , Técnicas de Sustitución del Gen , Hipocampo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Modelos Biológicos , Proteínas tau/metabolismo
10.
J Neuroinflammation ; 15(1): 91, 2018 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-29558961

RESUMEN

BACKGROUND: Apolipoprotein E (ApoE) is the major carrier protein that mediates the transport and delivery of cholesterol and other lipids in the brain. Three isoforms of ApoE (ApoE2, ApoE3, ApoE4) exist in humans, and their relative expression levels impact HIV-1 infection, HIV-1/AIDS disease progression, and cognitive decline associated with HIV-1-associated neurocognitive disorder. Because HIV-1 Tat, a viral protein essential for HIV-1 replication, can bind to low-density lipoprotein receptor-related protein 1 (LRP1) that controls ApoE uptake in the brain, we determined the extent to which different isoforms of ApoE affected Tat-mediated HIV-1 LTR transactivation. METHODS: Using U87MG glioblastoma cells expressing LTR-driven luciferase, we determined the extent to which LRP1 as well as ApoE2, ApoE3, and ApoE4 affected Tat-mediated HIV-1 LTR transactivation. RESULTS: A specific LRP1 antagonist and siRNA knockdown of LRP1 both restricted significantly Tat-mediated LTR transactivation. Of the three ApoEs, ApoE4 was the least potent and effective at preventing HIV-1 Tat internalization and at decreasing Tat-mediated HIV-1 LTR transactivation. Further, Tat-mediated LTR transactivation was attenuated by an ApoE mimetic peptide, and ApoE4-induced restriction of Tat-mediated LTR transactivation was potentiated by an ApoE4 structure modulator that changes ApoE4 into an ApoE3-like phenotype. CONCLUSIONS: These findings help explain observed differential effects of ApoEs on HIV-1 infectivity and the prevalence of HAND in people living with HIV-1 infection and suggest that ApoE mimetic peptides and ApoE4 structure modulator might be used as a therapeutic strategy against HIV-1 infection and associated neurocognitive disorders.


Asunto(s)
Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Duplicado del Terminal Largo de VIH/fisiología , Activación Transcripcional/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteína E4/farmacología , Línea Celular Tumoral , HDL-Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Duplicado del Terminal Largo de VIH/genética , Humanos , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/farmacología , Neuroblastoma/patología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Activación Transcripcional/efectos de los fármacos , Transfección
11.
Neuron ; 96(5): 1024-1032.e3, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29216449

RESUMEN

Accumulation and aggregation of amyloid-ß (Aß) in the brain is an initiating step in the pathogenesis of Alzheimer's disease (AD). The ε4 allele of apolipoprotein E (apoE) gene is the strongest genetic risk factor for late-onset AD. Although there is strong evidence showing that apoE4 enhances amyloid pathology, it is not clear what the critical stage(s) is during amyloid development in which apoE4 has the strongest impact. Using apoE inducible mouse models, we show that increased expression of astrocytic apoE4, but not apoE3, during the seeding stage of amyloid development enhanced amyloid deposition and neuritic dystrophy in amyloid model mice. ApoE4, but not apoE3, significantly increased brain Aß half-life measured by in vivo microdialysis. Furthermore, apoE4 expression increased whereas apoE3 reduced amyloid-related gliosis in the mouse brains. Together, our results demonstrate that apoE4 has the greatest impact on amyloid during the seeding stage, likely by perturbing Aß clearance and enhancing Aß aggregation.


Asunto(s)
Amiloidosis/patología , Apolipoproteína E4/farmacología , Enfermedad de Alzheimer/patología , Amiloidosis/genética , Animales , Apolipoproteína E3/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/patología , Técnicas de Sustitución del Gen , Gliosis/patología , Humanos , Ratones , Ratones Transgénicos , Neuritas/efectos de los fármacos , Neuritas/patología
12.
Neuroscience ; 301: 375-83, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26073697

RESUMEN

OBJECTIVE: The current study aimed to explore the effects of apolipoprotein e (ApoE) on intracellular calcium ([Ca(2+)]i) and apoptosis of neurons after mechanical injury in vitro. METHODS: A neuron mechanical injury model was established after primary neurons obtained from APOE knockout and wild-type (WT) mice, and four experimental groups were generated: Group-ApoE4, Group-ApoE3, Group-ApoE(-) and Group-WT. Recombinant ApoE4 and ApoE3 were added to Group-ApoE4 and Group-ApoE3 respectively, and Group-ApoE(-) and Group-WT were control groups. Intracellular calcium was labeled by fluo-3/AM and examined using laser scanning confocal microscope and flow cytometry, and the apoptosis of neurons was also evaluated. RESULTS: The intracellular calcium levels and apoptosis rates of mice neurons were significantly higher in Group-ApoE4 than in Group-ApoE3 and Group-WT after mechanical injury. However, without mechanical injury on neurons, no significant differences in intracellular calcium levels and apoptosis rates were found among all four experimental groups. The effects of ApoE4 on intracellular calcium levels and apoptosis rates of injured neurons were partly decreased by EGTA treatment. CONCLUSION: Compared with ApoE3-treatment and WT neurons, ApoE4 caused higher intracellular calcium levels and apoptosis rates of neurons after mechanical injury. This suggested APOE polymorphisms may affect neuron apoptosis after mechanical injury through different influences on intracellular calcium levels.


Asunto(s)
Apolipoproteínas E/fisiología , Apoptosis/genética , Calcio/metabolismo , Líquido Intracelular/metabolismo , Neuronas/metabolismo , Estimulación Física/efectos adversos , Análisis de Varianza , Animales , Apolipoproteína E3/farmacología , Apolipoproteína E4/farmacología , Apolipoproteínas E/deficiencia , Apoptosis/efectos de los fármacos , Corteza Cerebral/citología , Citometría de Flujo , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Estrés Mecánico , Factores de Tiempo
13.
ACS Nano ; 8(3): 2345-59, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24527692

RESUMEN

Amyloid-beta (Aß) accumulation in the brain is believed to play a central role in Alzheimer's disease (AD) pathogenesis, and the common late-onset form of AD is characterized by an overall impairment in Aß clearance. Therefore, development of nanomedicine that can facilitate Aß clearance represents a promising strategy for AD intervention. However, previous work of this kind was concentrated at the molecular level, and the disease-modifying effectiveness of such nanomedicine has not been investigated in clinically relevant biological systems. Here, we hypothesized that a biologically inspired nanostructure, apolipoprotein E3-reconstituted high density lipoprotein (ApoE3-rHDL), which presents high binding affinity to Aß, might serve as a novel nanomedicine for disease modification in AD by accelerating Aß clearance. Surface plasmon resonance, transmission electron microscopy, and co-immunoprecipitation analysis showed that ApoE3-rHDL demonstrated high binding affinity to both Aß monomer and oligomer. It also accelerated the microglial, astroglial, and liver cell degradation of Aß by facilitating the lysosomal transport. One hour after intravenous administration, about 0.4% ID/g of ApoE3-rHDL gained access to the brain. Four-week daily treatment with ApoE3-rHDL decreased Aß deposition, attenuated microgliosis, ameliorated neurologic changes, and rescued memory deficits in an AD animal model. The findings here provided the direct evidence of a biomimetic nanostructure crossing the blood-brain barrier, capturing Aß and facilitating its degradation by glial cells, indicating that ApoE3-rHDL might serve as a novel nanomedicine for disease modification in AD by accelerating Aß clearance, which also justified the concept that nanostructures with Aß-binding affinity might provide a novel nanoplatform for AD therapy.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/química , Apolipoproteína E3/química , Apolipoproteína E3/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Nanopartículas , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E3/metabolismo , Apolipoproteína E3/uso terapéutico , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Cinética , Lipoproteínas HDL/metabolismo , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Nanomedicina , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína
14.
FEBS Lett ; 587(2): 254-8, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23238080

RESUMEN

Apolipoprotein E (apoE) is predominantly synthesized by astrocytes in the brain. In this study, we investigated the role of apoE in astrocyte apoptosis. We demonstrated that apoE protects astrocytes from hypoxia-induced apoptosis in a dose-dependent manner. Glutamate release from astrocytic cultures is significantly lower from WT mice than from apoE knockout mice. Furthermore, the protective effect of apoE is mimicked by an NMDA receptor antagonist, MK-801. Finally, the apoE activator T0901317 significantly reduced the effect of glutamate-induced apoptosis of astrocytes. These results suggest that apoE protects astrocytes from hypoxia-induced apoptosis associated to NMDA receptor activation. Approaches that elevate apoE secretion in astrocytes might provide a novel strategy in the protection of neuronal ischemic injury.


Asunto(s)
Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Animales , Apolipoproteína E3/farmacología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Astrocitos/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Células Cultivadas , Maleato de Dizocilpina/farmacología , Técnicas de Inactivación de Genes , Ácido Glutámico/toxicidad , Hidrocarburos Fluorados/farmacología , Hipoxia Encefálica/metabolismo , Hipoxia Encefálica/patología , Ratones , Microscopía Electrónica de Transmisión , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Sulfonamidas/farmacología
15.
PLoS One ; 7(11): e50513, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209766

RESUMEN

OBJECTIVE: Postprandial hyperlipemia, characterized by increased circulating very low-density lipoproteins (VLDL) and circulating lipopolysaccharide (LPS), has been proposed as a mechanism of vascular injury. Our goal was to examine the interactions between postprandial lipoproteins, LPS, and apoE3 and apoE4 on monocyte activation. METHODS AND RESULTS: We showed that apoE3 complexed to phospholipid vesicles attenuates LPS-induced THP-1 monocyte cytokine expression, while apoE4 increases expression. ELISA revealed that apoE3 binds to LPS with higher affinity than apoE4. Electron paramagnetic resonance (EPR) spectroscopy of site-directed spin labels placed on specific amino acids of apoE3 showed that LPS interferes with conformational changes normally associated with lipid binding. Specifically, compared to apoE4, apoE bearing the E3-like R112→Ser mutation displays increased self association when exposed to LPS, consistent with a stronger apoE3-LPS interaction. Additionally, lipolysis of fasting VLDL from normal human donors attenuated LPS-induced TNFα secretion from monocytes to a greater extent than postprandial VLDL, an effect partially reversed by blocking apoE. This effect was reproduced using fasting VLDL lipolysis products from e3/e3 donors, but not from e4/e4 subjects, suggesting that apoE3 on fasting VLDL prevents LPS-induced inflammation more readily than apoE4. CONCLUSION: Postprandial apoE isoform and conformational changes associated with VLDL dramatically modulate vascular inflammation.


Asunto(s)
Apolipoproteínas E/química , Apolipoproteínas E/metabolismo , Lipólisis/efectos de los fármacos , Lipoproteínas VLDL/metabolismo , Monocitos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Adolescente , Adulto , Apolipoproteína E3/química , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/química , Apolipoproteína E4/metabolismo , Apolipoproteína E4/farmacología , Apolipoproteínas E/farmacología , Línea Celular , Espectroscopía de Resonancia por Spin del Electrón , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Lipopolisacáridos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/inmunología , Isoformas de Proteínas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
16.
Cell Rep ; 2(5): 1259-71, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23103162

RESUMEN

Arterial stiffening is a risk factor for cardiovascular disease, but how arteries stay supple is unknown. Here, we show that apolipoprotein E (apoE) and apoE-containing high-density lipoprotein (apoE-HDL) maintain arterial elasticity by suppressing the expression of extracellular matrix genes. ApoE interrupts a mechanically driven feed-forward loop that increases the expression of collagen-I, fibronectin, and lysyl oxidase in response to substratum stiffening. These effects are independent of the apoE lipid-binding domain and transduced by Cox2 and miR-145. Arterial stiffness is increased in apoE null mice. This stiffening can be reduced by administration of the lysyl oxidase inhibitor BAPN, and BAPN treatment attenuates atherosclerosis despite highly elevated cholesterol. Macrophage abundance in lesions is reduced by BAPN in vivo, and monocyte/macrophage adhesion is reduced by substratum softening in vitro. We conclude that apoE and apoE-containing HDL promote healthy arterial biomechanics and that this confers protection from cardiovascular disease independent of the established apoE-HDL effect on cholesterol.


Asunto(s)
Apolipoproteínas E/metabolismo , HDL-Colesterol/farmacología , Matriz Extracelular/metabolismo , Aminopropionitrilo/farmacología , Aminopropionitrilo/uso terapéutico , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas , Colágeno Tipo I/metabolismo , Ciclooxigenasa 2/metabolismo , Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Expresión Génica , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Rigidez Vascular/efectos de los fármacos
17.
J Biol Chem ; 287(19): 15947-58, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22427674

RESUMEN

Synaptic loss is the earliest pathological change in Alzheimer disease (AD) and is the pathological change most directly correlated with the degree of dementia. ApoE4 is the major genetic risk factor for the age-dependent form of AD, which accounts for 95% of cases. Here we show that in synaptic networks formed from primary hippocampal neurons in culture, apoE3, but not apoE4, prevents the loss of synaptic networks produced by amyloid ß oligomers (amylospheroids). Specific activators of PKCε, such as 8-(2-(2-pentyl-cyclopropylmethyl)-cyclopropyl)-octanoic acid methyl ester and bryostatin 1, protected against synaptic loss by amylospheroids, whereas PKCε inhibitors blocked this synaptic protection and also blocked the protection by apoE3. Blocking LRP1, an apoE receptor on the neuronal membrane, also blocked the protection by apoE. ApoE3, but not apoE4, induced the synthesis of PKCε mRNA and expression of the PKCε protein. Amyloid ß specifically blocked the expression of PKCε but had no effect on other isoforms. These results suggest that protection against synaptic loss by apoE is mediated by a novel intracellular PKCε pathway. This apoE pathway may account for much of the protective effect of apoE and reduced risk for the age-dependent form of AD. This finding supports the potential efficacy of newly developed therapeutics for AD.


Asunto(s)
Apolipoproteína E3/farmacología , Apolipoproteína E4/farmacología , Neuronas/efectos de los fármacos , Proteína Quinasa C-epsilon/metabolismo , Sinapsis/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Western Blotting , Brioestatinas/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colesterol/farmacología , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Neuronas/metabolismo , Neuronas/patología , Proteína Quinasa C-epsilon/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/metabolismo , Sinapsis/patología
18.
J Alzheimers Dis ; 21(2): 403-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20555142

RESUMEN

The epsilon4 allele of apolipoprotein E (APOE) is currently the major genetic risk factor identified for Alzheimer's disease (AD). Previous in vivo data from our laboratory has demonstrated that amyloid-beta (Abeta) is rapidly removed from the plasma by the liver and kidney and that the rate of its clearance is affected by ApoE in C57BL/6J and APOE-/- mice. To expand upon these findings, we assessed the peripheral clearance of human synthetic Abeta42 in APOE epsilon2, epsilon3, and epsilon4 knock-in and APOE knock-out mice injected with lipidated recombinant apoE2, E3, and E4 protein. Our results show that APOE does influence the rate at which the mice are able to clear Abeta42 from their bloodstream. Both APOE epsilon4 mice and APOE knock-out mice treated with lipidated recombinant apoE4 demonstrated increased retention of plasma Abeta42 over time compared to APOE epsilon2/APOE knock-out rE2 and APOE epsilon3/APOE knock-out rE3 mice. These findings suggest that the peripheral clearance of Abeta42 is significantly altered by APOE genotype. Given that APOE epsilon4 is a risk factor for AD, then these novel findings provide some insight into the role of ApoE isoforms on the peripheral clearance of Abeta which may impact on clearance from the brain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacocinética , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Fragmentos de Péptidos/farmacocinética , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/sangre , Animales , Apolipoproteína E2/farmacología , Apolipoproteína E3/farmacología , Apolipoproteína E4/farmacología , Encéfalo/metabolismo , Técnicas de Sustitución del Gen , Genotipo , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/sangre , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Distribución Tisular
19.
Protein Expr Purif ; 68(1): 7-11, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19545633

RESUMEN

Apolipoprotein E3 (ApoE3) is an important apolipoprotein in plasma and plays a critical role in lipid transport and cholesterol homeostasis. As the only natural source of this protein, human blood cannot provide large-scale ApoE3 for research and applications. Therefore, in our study, a Pichia pastoris expression system was first used to obtain a high-level expression of secreted, recombinant human ApoE3 (rhApoE3). The full-length sequence encoding ApoE3, gained by RT-PCR, was inserted into the pPICZalphaC vector and transformed into P. pastoris strain X33, and then the high expression transformants with zeocin resistance were obtained. The growth conditions of the transformant strains were optimized in 50ml conical tubes including pH and inducing time. After induction with methanol, the expression level of rhApoE3 was 120 mg/L in 80 L fermentor. RhApoE3 was purified more than 94% purity using SP Sepharose ion exchange chromatography and source 30RPC. A preliminary biochemical characterization of purified rhApoE3 was performed by analyzing the ability of inhibiting PDGF-induced proliferation of rat coronary artery smooth muscle cells (SMCs), and the results demonstrated that the function of purified rhApoE3 was similar to natural human ApoE3.


Asunto(s)
Apolipoproteína E3/química , Apolipoproteína E3/metabolismo , Técnicas de Cultivo de Célula/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/farmacología , Reactores Biológicos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Clonación Molecular/métodos , Electroforesis en Gel de Poliacrilamida , Fermentación , Humanos , Concentración de Iones de Hidrógeno , Miocitos del Músculo Liso , Pichia/genética , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Tiempo
20.
Neuroscience ; 162(2): 282-91, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19414061

RESUMEN

Co-cultures of 3T3-L1 adipocytes with neurons from the rat dorsal root ganglia (DRG) showed enhanced neuritogenesis and synaptogenesis. Microarray analysis for upregulated genes in adipocyte/DRG co-cultures currently points to apolipoproteins D and E (ApoD, ApoE) as influential proteins. We therefore tested adipocyte-secreted cholesterol and the carrier proteins ApoD and ApoE3. Cholesterol, ApoD, and ApoE3 each increased neurite outgrowth and upregulated the expression of presynaptic synaptophysin and synaptotagmin, as well as the postsynaptic density protein 95. The neurotrophic effects of ApoD and ApoE3 were associated with an increased expression of the low-density lipoprotein receptor and apolipoprotein E receptor 2. Simultaneous treatment with receptor-associated protein, an apolipoprotein receptor antagonist, inhibited the neurotrophic function of both apolipoproteins. The application of ApoD, ApoE3, and cholesterol to DRG cell cultures corresponded with increased expression of the chemokine stromal cell-derived factor 1 and its receptor CXC chemokine receptor 4 (CXCR4). Surprisingly, the inhibition of CXCR4 by the antagonistic drug AMD3100 decreased the apolipoprotein/cholesterol dependent neurotrophic effects. We thus assume that apolipoprotein-induced neuritogenesis in DRG cells interferes with CXCR4 signaling, and that adipocyte-derived apolipoproteins might be helpful in nerve repair.


Asunto(s)
Apolipoproteína E3/fisiología , Apolipoproteínas D/fisiología , Ganglios Espinales/citología , Neuronas/fisiología , Sinapsis/fisiología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipoquinas/biosíntesis , Animales , Apolipoproteína E3/farmacología , Apolipoproteínas D/farmacología , Bencilaminas , Células Cultivadas , Quimiocina CXCL12/biosíntesis , Colesterol/farmacología , Colesterol/fisiología , Técnicas de Cocultivo , Ciclamas , Homólogo 4 de la Proteína Discs Large , Compuestos Heterocíclicos/farmacología , Péptidos y Proteínas de Señalización Intracelular , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/farmacología , Proteínas de la Membrana/biosíntesis , Neuritas/fisiología , Neuronas/efectos de los fármacos , Ratas , Ratas Endogámicas WF , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/biosíntesis , Receptores de Lipoproteína/antagonistas & inhibidores , Receptores de Lipoproteína/metabolismo , Sinapsis/efectos de los fármacos , Sinaptofisina/biosíntesis , Sinaptotagminas/biosíntesis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...