Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
1.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806327

ABSTRACT

Recent evidence suggests that I2-imidazoline ligands have neuroprotective properties in animal models of neurodegeneration, such as Alzheimer's disease (AD). We recently demonstrated that the I2-ligand BU224 reversed memory impairments in AD transgenic mice and this effect was not because of reductions in amyloid-ß (Aß) deposition. In this study, our aim was to determine the therapeutic potential of the powerful analgesic I2-imidazoline ligand CR4056 in the 5xFAD model of AD, since this ligand has been proven to be safely tolerated in humans. Sub-chronic oral administration of CR4056 (30 mg/kg for 10 days) led to an improvement in recognition memory in 6-month-old 5xFAD mice, but not in wild-type littermates, without affecting Aß levels or deposition. Our results also revealed a change in the profile of microglia by CR4056, resulting in a suppression of pro-inflammatory activated microglia, but increased the density of astrocytes and the expression of ApoE, which is mainly produced by these glial cells. In addition, CR4056 restored fibrinogen extravasation, affecting the distribution of markers of astrocytic end feet in blood vessels. Therefore, these results suggest that CR4056 protects against Aß-mediated neuroinflammation and vascular damage, and offers therapeutic potential at any stage of AD.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Imidazoles , Imidazolines , Quinazolines , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Imidazoles/pharmacology , Imidazolines/metabolism , Ligands , Mice , Mice, Transgenic , Quinazolines/pharmacology
2.
Nat Neurosci ; 24(6): 786-798, 2021 06.
Article in English | MEDLINE | ID: mdl-33958804

ABSTRACT

Selective neurodegeneration is a critical causal factor in Alzheimer's disease (AD); however, the mechanisms that lead some neurons to perish, whereas others remain resilient, are unknown. We sought potential drivers of this selective vulnerability using single-nucleus RNA sequencing and discovered that ApoE expression level is a substantial driver of neuronal variability. Strikingly, neuronal expression of ApoE-which has a robust genetic linkage to AD-correlated strongly, on a cell-by-cell basis, with immune response pathways in neurons in the brains of wild-type mice, human ApoE knock-in mice and humans with or without AD. Elimination or over-expression of neuronal ApoE revealed a causal relationship among ApoE expression, neuronal MHC-I expression, tau pathology and neurodegeneration. Functional reduction of MHC-I ameliorated tau pathology in ApoE4-expressing primary neurons and in mouse hippocampi expressing pathological tau. These findings suggest a mechanism linking neuronal ApoE expression to MHC-I expression and, subsequently, to tau pathology and selective neurodegeneration.


Subject(s)
Alzheimer Disease/metabolism , Apolipoproteins E/biosynthesis , Histocompatibility Antigens Class I/biosynthesis , Neurons/metabolism , Up-Regulation/physiology , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Apolipoproteins E/genetics , Cells, Cultured , Databases, Genetic/trends , Female , Gene Expression , Gene Knock-In Techniques/methods , Histocompatibility Antigens Class I/genetics , Humans , Male , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/pathology
3.
Toxicology ; 432: 152381, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31981724

ABSTRACT

Chronic glutamate excitotoxicity has been thought to be involved in numerous neurodegenerative disorders. A small but significant loss of membrane cholesterol has been reported following a short stimulation of ionotropic glutamate receptors (iGluRs). We investigated the alteration of brain cholesterol following chronic glutamate treatment. The alteration of cholesterol levels was evaluated in the hippocampus from the adult rats that received the subcutaneous injection with monosodium l-glutamate at 1, 3, 5, and 7 days of age. The regulation of CYP46A1, LXRα, and ApoE levels were assayed following subtoxic glutamate treatment in SH-SY5Y cells as well as HT-22 cells lacking iGluRs. The ratio of 24S-hydroxycholesterol to cholesterol was elevated in the adult rats exposed to monosodium l-glutamate before the weaning age, compared to the control. The blockers of NMDA receptor (MK801) and mGluR5 (MPEP) attenuated the glutamate-induced loss of cholesterol and elevation of 24S-hydroxycholesterol level in SH-SY5Y cells. The induction of the mRNA levels of CYP46A1, LXRα, and ApoE by glutamate was observed in both SH-SY5Y cells and HT-22 cells; additionally, MK801 and MPEP attenuated the increases in these genes in SH-SY5Y cells. The increase in the binding of LXRα proteins with ApoE promoter following glutamate treatment was attenuated by MK801. The luciferase assay indicated the binding of CREB protein with CYP46A1 promoter, and the glutamate-induced CREB expression was inhibited by MK801. The results suggest that glutamate, the major excitatory neurotransmitter, may affect the metabolism and redistribution of cholesterol in the neuronal cells via its specific receptors during chronic exposure.


Subject(s)
Apolipoproteins E/biosynthesis , Brain Chemistry/drug effects , Cholesterol 24-Hydroxylase/biosynthesis , Cholesterol/metabolism , Sodium Glutamate/pharmacology , Animals , Cell Line , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Homeostasis , Liver X Receptors/antagonists & inhibitors , Maze Learning/drug effects , Rats , Rats, Wistar , Receptors, Glutamate/drug effects , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Up-Regulation/drug effects
4.
Neurotox Res ; 37(1): 227-237, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31654382

ABSTRACT

The synthetic peptide p-BTX-I is based on the native peptide (formed by glutamic acid, valine and tryptophan) isolated from Bothrops atrox venom. We have previously demonstrated its neuroprotective and neurotrophic properties in PC12 cells treated with the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium (MPP+). Now, we have investigated the neuroprotective effects and mechanisms of p-BTX-I against the toxicity of acrolein in PC12 cells. Studies have demonstrated that acrolein might play an important role in the etiology of Alzheimer's disease (AD), which is characterized by neuronal and synaptic loss. Our results showed that not only acrolein reduced cell differentiation and cell viability, but also altered the expression of markers of synaptic communication (synapsin I), energy metabolism (AMPK-α, Sirt I and glucose uptake), and cytoskeleton (ß-III-tubulin). Treatment with p-BTX-I increased the percentage of differentiation in cells treated with acrolein and significantly attenuated cell viability loss, besides counteracting the negative effects of acrolein on synapsin I, AMPK-α, Sirt I, glucose uptake, and ß-III-tubulin. Additionally, p-BTX-I alone increased the expression of apolipoprotein E (apoE) gene, associated with the proteolytic degradation of ß-amyloid peptide aggregates, a hallmark of AD. Taken together, these findings demonstrate that p-BTX-I protects against acrolein-induced neurotoxicity and might be a tool for the development of novel drugs for the treatment of neurodegenerative diseases.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Acrolein/antagonists & inhibitors , Energy Metabolism/drug effects , Glucose/metabolism , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Sirtuin 1/biosynthesis , Synapsins/biosynthesis , Tubulin/biosynthesis , Acrolein/toxicity , Animals , Apolipoproteins E/biosynthesis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , PC12 Cells , Peptides/pharmacology , Rats
5.
Arterioscler Thromb Vasc Biol ; 40(1): 128-144, 2020 01.
Article in English | MEDLINE | ID: mdl-31665905

ABSTRACT

OBJECTIVE: The ε4 allele of the APOE gene (APOE4) is the strongest genetic risk factor for Alzheimer disease when compared with the common ε3 allele. Although there has been significant progress in understanding how apoE4 (apolipoprotein E4) drives amyloid pathology, its effects on amyloid-independent pathways, in particular cerebrovascular integrity and function, are less clear. Approach and Results: Here, we show that brain pericytes, the mural cells of the capillary walls, differentially modulate endothelial cell phenotype in an apoE isoform-dependent manner. Extracellular matrix protein induction, tube-like structure formation, and barrier formation were lower with endothelial cells cocultured with pericytes isolated from apoE4-targeted replacement (TR) mice compared with those from apoE3-TR mice. Importantly, aged apoE4-targeted replacement mice had decreased extracellular matrix protein expression and increased plasma protein leakages compared with apoE3-TR mice. CONCLUSIONS: ApoE4 impairs pericyte-mediated basement membrane formation, potentially contributing to the cerebrovascular effects of apoE4.


Subject(s)
Alzheimer Disease/genetics , Apolipoproteins E/genetics , Basement Membrane/metabolism , Brain/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation , Pericytes/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Apolipoproteins E/biosynthesis , Basement Membrane/pathology , Brain/pathology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Mice, Inbred C57BL , Pericytes/pathology , Protein Isoforms
6.
Ocul Surf ; 17(4): 777-786, 2019 10.
Article in English | MEDLINE | ID: mdl-31201956

ABSTRACT

PURPOSE: To investigate the pathological changes of the meibomian gland (MG) and ocular surface in Apolipoprotein E knockout (ApoE-/-) mice and to investigate the association of meibomian gland dysfunction (MGD) with hyperlipidemia. METHODS: Total plasma cholesterol was measured in different ages of ApoE-/- and wild type (WT) mice, whilst the ocular surfaces were observed by slit-lamp biomicroscopy. MG sections were subjected to H&E staining, Oil Red O staining, TUNEL assay and immunostaining. Quantitate RT-PCR and Western blot analyses were performed to detect the relative gene expression in MGs. The 5-month-old ApoE-/- mice were administered with rosiglitazone or GW9662 + rosiglitazone via oral gavage for 2 months to determine their effect on MG pathological change. RESULTS: We found eyelid abnormality, MG dropout, abnormal MG acinar morphology, dilated MG duct and plugging of the MG orifice in ApoE-/- mice. MG acini in ApoE-/- mice showed exaggerated lipid accumulation. Abnormal keratinization increased in MG duct, accompanied with decreased proliferation and increased apoptosis in ApoE-/- mice. Inflammatory cells infiltrated into the surrounding microenvironment of MG acini, and the NF-κB signaling pathway was activated in MG acinar cells. Oxidative stress was evident in MG acinar cells of ApoE-/- mice. Further investigation showed downregulation of PPAR-γ in MG acinar cells of ApoE-/- mice. PPAR-γ agonist rosiglitazone treatment reduced the morbidity of eyelid, as well as corneal pathological changes and MG inflammation in ApoE-/- mice. CONCLUSION: MGD and hyperlipidemia are closely associated in ApoE-/- mice, which represent a new model to study the pathophysiology of MGD related to dyslipidemia.


Subject(s)
Apolipoproteins E/genetics , Gene Expression Regulation , Hyperlipidemias/complications , Meibomian Gland Dysfunction/etiology , Meibomian Glands/pathology , RNA/genetics , Animals , Apolipoproteins E/biosynthesis , Apolipoproteins E/blood , Blotting, Western , Disease Models, Animal , Hyperlipidemias/metabolism , Male , Meibomian Gland Dysfunction/diagnosis , Meibomian Gland Dysfunction/metabolism , Meibomian Glands/metabolism , Mice , Mice, Knockout , Rabbits
7.
Mol Cell Endocrinol ; 490: 21-27, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30953750

ABSTRACT

Apolipoprotein E (APOE) deficient mice exhibit unexplained hypercorticosteronemia. Given that APOE is also produced locally within the adrenals, we evaluated the effect of adrenal-specific APOE deficiency on the glucocorticoid function. Hereto, one adrenal containing or lacking APOE was transplanted into adrenalectomized wild-type mice. Adrenal APOE deficiency did not impact adrenal total cholesterol levels. Importantly, the ability of the two adrenal types to produce glucocorticoids was also not different as judged from the similar plasma corticosterone levels. Adrenal mRNA expression levels of HMG-CoA reductase and the LDL receptor were decreased by respectively 72% (p < 0.01) and 65% (p = 0.07), suggesting that cholesterol acquisition pathways were inhibited to possibly compensate the lack of APOE. In support, a parallel increase in the expression level of the cholesterol accumulation-associated ER stress marker CHOP was detected (+117%; p < 0.05). In conclusion, our studies show that elimination of adrenocortical APOE production does not impact glucocorticoid output in wild-type mice.


Subject(s)
Adrenal Cortex/metabolism , Apolipoproteins E/biosynthesis , Glucocorticoids/pharmacology , Adrenal Cortex/drug effects , Animals , Apolipoproteins E/deficiency , Cholesterol/metabolism , Endoplasmic Reticulum Stress/drug effects , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
J Cell Physiol ; 233(9): 6925-6943, 2018 09.
Article in English | MEDLINE | ID: mdl-29323721

ABSTRACT

In the central nervous system, cholesterol is critical to maintain membrane plasticity, cellular function, and synaptic integrity. In recent years, much attention was focused on the role of cholesterol in brain since a breakdown of cholesterol metabolism has been associated with different diseases. Brain-derived neurotrophic factor (BDNF) was previously reported to elicit cholesterol biosynthesis and promote the accumulation of presynaptic proteins in cholesterol-rich lipid rafts, but no data are available on its ability to modulate physiological mechanisms involved in cholesterol homeostasis. Major aim of this research was to investigate whether BDNF influences cholesterol homeostasis, focusing on the effect of the neurotrophin on Apolipoprotein E (ApoE) synthesis, cholesterol efflux from astrocytes and cholesterol incorporation into neurons. Our results show that BDNF significantly stimulates cholesterol efflux by astrocytes, as well as ATP binding cassette A1 (ABCA1) transporter and ApoE expression. Conversely, cholesterol uptake in neurons was downregulated by BDNF. This effect was associated with the increase of Liver X Receptor (LXR)-beta expression in neuron exposed to BDNF. The level of apoptosis markers, that is, cleaved caspase 3 and poly ADP ribose polymerase (PARP), was found increased in neurons treated with high cholesterol, but significantly lower when the cells were exposed to cholesterol in the presence of BDNF, thus suggesting a neuroprotective role of the neurotrophin, likely through its reducing effect of neuronal cholesterol uptake. Interestingly, cholesterol stimulates BDNF production by neurons. Overall, our findings evidenced a novel role of BDNF in the modulation of ApoE and cholesterol homeostasis in glial and neuronal cells.


Subject(s)
Apolipoproteins E/biosynthesis , Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cholesterol/metabolism , Homeostasis/drug effects , Neurons/metabolism , ATP Binding Cassette Transporter 1/metabolism , Apoptosis/drug effects , Astrocytes/drug effects , Cell Line, Tumor , Humans , Liver X Receptors/metabolism , MAP Kinase Signaling System/drug effects , Middle Aged , Models, Biological , Neurons/drug effects , Prefrontal Cortex/metabolism
9.
Sci Rep ; 7(1): 2495, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566716

ABSTRACT

Direct acting antivirals against hepatitis C virus (HCV) have markedly improved cure rates in the past few years. However, they are expensive, with only few targeting host cell factors, and affecting virus assembly and release. Huh7.5 cells infected with a JFH-1 clone of HCV were treated with two different glycogen synthase kinase (GSK3)-ß inhibitors; AR-A014418 and lithium chloride. Intra- and extracellular HCV virions and specific infectivity was determined using real-time RT-PCR and TCID50, and changes in lipid production were identified by enzyme-linked immunoassay and mass spectrometry analyses. Similarly, effect on two HCV replicon cells were identified by the luciferase activity. Although there was limited effect on virus replication in Huh7.5 cells and replicons, Huh7.5 cells treated with GSK3ß inhibitors produced significantly less viral particles in comparison to untreated cells. In addition, the treated cells synthesized significantly lower amounts of ApoB and trapped the ApoE lipoproteins in the cells. In conclusion, our study suggests that GSK3ß plays a pivotal role in HCV virion assembly and release mediated in part through inhibition of apolipoprotein synthesis.


Subject(s)
Glycogen Synthase Kinase 3 beta/genetics , Hepatitis C/drug therapy , Lipid Metabolism/drug effects , Virus Replication/drug effects , Apolipoproteins B/biosynthesis , Apolipoproteins B/genetics , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Cell Line , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hepacivirus/drug effects , Hepacivirus/pathogenicity , Hepatitis C/genetics , Hepatitis C/virology , Humans , Lithium Chloride/pharmacology , Thiazoles/pharmacology , Urea/analogs & derivatives , Urea/pharmacology , Virion/drug effects , Virion/genetics
10.
Invest Ophthalmol Vis Sci ; 58(7): 3073-3085, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28632844

ABSTRACT

PURPOSE: Complement activation is implicated in the pathogenesis of age-related macular degeneration (AMD). Apolipoprotein E (ApoE) and complement activation products such as membrane attack complex (MAC) are present in eyes of individuals with AMD. Herein, we investigated the effect of complement activation on induction of ApoE accumulation in human retinal pigment epithelial (RPE) cells. METHODS: Cultured human RPE cells were primed with a complement-fixing antibody followed by treatment with C1q-depleted (C1q-Dep) human serum to elicit alternative pathway complement activation. Controls included anti-C5 antibody-treated serum and heat-inactivated C1q-Dep. Total protein was determined on RPE cell extracts, conditioned media, and extracellular matrix (ECM) by Western blot. ApoE and MAC colocalization was assessed on cultured RPE cells and human eyes by immunofluorescent stain. ApoE mRNA expression was evaluated by quantitative PCR (qPCR). RESULTS: Complement challenge upregulated cell-associated ApoE, but not apolipoprotein A1. ApoE accumulation was blocked by anti-C5 antibody and enhanced by repetitive complement challenge. ApoE mRNA levels were not affected by complement challenge. ApoE was frequently colocalized with MAC in complement-treated cells and drusen from human eyes. ApoE was released into complement-treated conditioned media after a single complement challenge and accumulated on ECM after repetitive complement challenge. CONCLUSIONS: Complement challenge induces time-dependent ApoE accumulation in RPE cells. An understanding of the mechanisms by which complement affects RPE ApoE accumulation may help to better explain drusen composition, and provide insights into potential therapeutic targets.


Subject(s)
Apolipoproteins E/genetics , Complement Activation/physiology , Gene Expression Regulation , Macular Degeneration/genetics , RNA/genetics , Retinal Pigment Epithelium/metabolism , Apolipoproteins E/biosynthesis , Blotting, Western , Cells, Cultured , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mass Spectrometry , Middle Aged , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/pathology
11.
J Neuropathol Exp Neurol ; 76(4): 270-288, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28340083

ABSTRACT

The term "aging-related tau astrogliopathy" (ARTAG) describes pathological accumulation of abnormally phosphorylated tau protein in astrocytes. We evaluated the correlates of ARTAG types (i.e., subpial, subependymal, white and gray matter, and perivascular) in different neuroanatomical regions. Clinical, neuropathological, and genetic (eg, APOE ε4 allele, MAPT H1/H2 haplotype) data from 628 postmortem brains from subjects were investigated; most of the patients had been longitudinally followed at the University of Pennsylvania. We found that (i) the amygdala is a hotspot for all ARTAG types; (ii) age at death, male sex, and presence of primary frontotemporal lobar degeneration (FTLD) tauopathy are significantly associated with ARTAG; (iii) age at death, greater degree of brain atrophy, ventricular enlargement, and Alzheimer disease (AD)-related variables are associated with subpial, white matter, and perivascular ARTAG types; (iv) AD-related variables are associated particularly with lobar white matter ARTAG; and (v) gray matter ARTAG in primary FTLD-tauopathies appears in areas without neuronal tau pathology. We provide a reference map of ARTAG types and propose at least 5 constellations of ARTAG. Furthermore, we propose a conceptual link between primary FTLD-tauopathy and ARTAG-related astrocytic tau pathologies. Our observations serve as a basis for etiological stratification and definition of progression patterns of ARTAG.


Subject(s)
Aging/pathology , Astrocytes/pathology , Brain/pathology , Neurodegenerative Diseases/pathology , Neuroglia/pathology , Tauopathies/pathology , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Atrophy , Female , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , Humans , Longitudinal Studies , Male , Sex Factors , tau Proteins/biosynthesis , tau Proteins/genetics
12.
Stroke ; 48(2): 459-467, 2017 02.
Article in English | MEDLINE | ID: mdl-28028143

ABSTRACT

BACKGROUND AND PURPOSE: ATP-binding cassette transporter A1 (ABCA1) is a major reverse cholesterol transporter and plays critical role in the formation of brain high-density lipoprotein (HDL) cholesterol. Apolipoprotein E (ApoE) is the most abundant apolipoprotein and transports cholesterol into cells in brain. ABCA1 and ApoE are upregulated by liver-X receptors. Activation of liver-X receptors has neurorestorative benefit for stroke. The current study investigates whether ABCA1/ApoE/HDL pathway mediates GW3965, a synthetic dual liver-X receptor agonist, induced neurorestoration after stroke. METHODS: Middle-aged male specific brain ABCA1-deficient (ABCA1-B/-B) and floxed-control (ABCA1fl/fl) mice were subjected to distal middle-cerebral artery occlusion (dMCAo) and gavaged with saline or GW3965 (10 mg/kg) or intracerebral infusion of artificial cerebrospinal fluid or human plasma HDL3 in ABCA1-B/-B stroke mice, starting 24 hours after dMCAo and daily until euthanization 14 days after dMCAo. RESULTS: No differences in the blood level of total cholesterol and triglyceride and lesion volume were found among the groups. Compared with ABCA1fl/fl ischemic mice, ABCA1-B/-B ischemic mice exhibited impairment functional outcome and decreased ABCA1/ApoE expression and decreased gray/white matter densities in the ischemic boundary zone 14 days after dMCAo. GW3965 treatment of ABCA1fl/fl ischemic mice led to increased brain ABCA1/ApoE expression, concomitantly to increased blood HDL, gray/white matter densities and oligodendrocyte progenitor cell numbers in the ischemic boundary zone, as well as improved functional outcome 14 days after dMCAo. GW3965 treatment had negligible beneficial effects in ABCA1-B/-B ischemic mice. However, intracerebral infusion of human plasma HDL3 significantly attenuated ABCA1-B/-B-induced deficits. In vitro, GW3965 treatment (5 µM) increased ABCA1/synaptophysin level and neurite/axonal outgrowth in primary cortical neurons derived from ABCA1fl/fl embryos, but not in neurons derived from ABCA1-B/-B embryos. HDL treatment (80 µg/mL) attenuated the reduction of neurite/axonal outgrowth in neurons derived from ABCA1-B/-B embryos. CONCLUSIONS: ABCA1/ApoE/HDL pathway, at least partially, contributes to GW3965-induced neurorestoration after stroke.


Subject(s)
ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter 1/deficiency , Apolipoproteins E/biosynthesis , Benzoates/administration & dosage , Benzylamines/administration & dosage , Lipoproteins, HDL/biosynthesis , Stroke/drug therapy , Stroke/metabolism , Animals , Cholesterol, HDL/administration & dosage , Humans , Infusions, Intraventricular , Male , Mice , Mice, Knockout , Random Allocation , Signal Transduction/drug effects , Signal Transduction/physiology , Stroke/pathology
13.
Stem Cells ; 33(12): 3643-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26417967

ABSTRACT

Epigenetic alterations play a central role in the control of normal and malignant blood cell development. We demonstrate here that expression of a truncated DNA methyltransferase 3B isoform DNMT3B7, which has been shown to alter cellular epigenetic patterns, decreases the overall number of hematopoietic stem and progenitor cells (HSPCs), and markedly diminishes blood cell reconstitution within the female hormonal microenvironment. Gene expression profiling of HSPCs isolated from DNMT3B7 transgenic embryos identified Apolipoprotein E (Apoe) as overexpressed. The CpG island controlling Apoe expression had lower levels of modified cytosines in DNMT3B7 transgenic HSPCs, corresponding with the observed increase in gene expression. Furthermore, we observed that spleens and bone marrows of female mice transplanted with DNMT3B7 transgenic HSPCs express very high levels of Apoe. Finally, the introduction of Apoe-overexpressing HSPCs into male recipients decreased bone marrow engraftment, recapitulating our original observations in female recipients. Our work reveals a dynamic interplay between the intrinsic epigenetic changes in HSPCs and extrinsic endocrine factors acting on these cells to regulate the efficiency of HSPC engraftment and reconstitution. We have identified a novel mechanism by which gender-specific hormones modulate HSPC function, which could serve as a target for augmenting hematopoiesis in cases with limited HSC functionality.


Subject(s)
Apolipoproteins E/biosynthesis , CpG Islands/physiology , Epigenesis, Genetic/physiology , Hematopoiesis/physiology , Sex Characteristics , Animals , Apolipoproteins E/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Male , Mice , Mice, Knockout
14.
J Lipid Res ; 56(3): 515-525, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25593327

ABSTRACT

Cells produce two cholesteryl ester transfer protein (CETP) isoforms, full-length and a shorter variant produced by alternative splicing. Blocking synthesis of both isoforms disrupts lipid metabolism and storage. To further define the role of CETP in cellular lipid metabolism, we stably overexpressed full-length CETP in SW872 cells. These CETP(+) cells had several-fold higher intracellular CETP and accumulated 50% less TG due to a 26% decrease in TG synthesis and 2.5-fold higher TG turnover rate. Reduced TG synthesis was due to decreased fatty acid uptake and impaired conversion of diglyceride to TG even though diacylglycerol acyltransferase activity was normal. Sterol-regulatory element binding protein 1 mRNA levels were normal, and although PPARγ expression was reduced, the expression of several of its target genes including adipocyte triglyceride lipase, FASN, and APOE was normal. CETP(+) cells contained smaller lipid droplets, consistent with their higher levels of perilipin protein family (PLIN) 3 compared with PLIN1 and PLIN2. Intracellular CETP was mostly associated with the endoplasmic reticulum, although CETP near lipid droplets poorly colocalized with this membrane. A small pool of CETP resided in the cytoplasm, and a subfraction coisolated with lipid droplets. These data show that overexpression of full-length CETP disrupts lipid homeostasis resulting in the formation of smaller, more metabolically active lipid droplets.


Subject(s)
Cholesterol Ester Transfer Proteins/metabolism , Cytoplasm/metabolism , Lipid Metabolism/physiology , Triglycerides/metabolism , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Cell Line, Tumor , Cholesterol Ester Transfer Proteins/genetics , Cytoplasm/genetics , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation/physiology , Humans , PPAR gamma/biosynthesis , PPAR gamma/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/genetics
15.
J Lipid Res ; 55(10): 2073-81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25183802

ABSTRACT

Lipoprotein glomerulopathy (LPG) is a renal disease often accompanied by dyslipidemia and increased serum apoE levels. apoESendai (Arg145Pro), a rare mutant based on the apoE3 sequence carrying an apoE2 charge, causes LPG in humans and transgenic mice, but its effects on the artery wall are unknown. Macrophage expression of apoESendai may also directly influence renal and arterial homeostasis. We investigated the effects of macrophage-expressed apoESendai in apoE(-/-) mice with or without LDL receptor (LDLR). Murine bone marrow transduced to express apoE2, apoE3, or apoESendai was transplanted into lethally irradiated mice. Macrophage apoESendai expression reduced aortic lesion size and inflammation by 32 and 28%, respectively, compared with apoE2 in apoE(-/-) recipients. No differences in lesion size or inflammation were found between apoESendai and apoE3 in apoE(-/-) recipients. Macrophage apoESendai expression also reduced aortic lesion size by 18% and inflammation by 29% compared with apoE2 in apoE(-/-)/LDLR(-/-) recipients. Glomerular lesions compatible with LPG with increased mesangial matrix, extracellular lipid accumulation, and focal mesangiolysis were only observed in apoE(-/-)/LDLR(-/-) mice expressing apoESendai. Thus, macrophage expression of apoESendai protects against atherosclerosis while causing lipoprotein glomerulopathy. This is the first demonstration of an apoprotein variant having opposing effects on vascular and renal homeostasis.


Subject(s)
Apolipoproteins E/biosynthesis , Atherosclerosis/metabolism , Glomerular Mesangium/metabolism , Hyperlipidemias/metabolism , Kidney Diseases/metabolism , Macrophages/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Glomerular Mesangium/pathology , Hyperlipidemias/genetics , Hyperlipidemias/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Macrophages/pathology , Mice , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism
16.
J Stroke Cerebrovasc Dis ; 23(9): 2350-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25194744

ABSTRACT

BACKGROUND: Telmisartan is a unique angiotensin II type 1 receptor blocker with a partial peroxisome proliferator-activated receptor-γ (PPARγ) agonistic property to exert not only antihypertensive effect but also antimetabolic syndrome effect. METHODS: We examined the long-term effect of telmisartan on cholesterol transport-related proteins (low-density lipoprotein receptor [LDL-R]/apolipoprotein E [ApoE]) and microtubule-associated proteins 2 (MAP2) in the brains of stroke resistant spontaneously hypertensive rats (SHR-SRs), which were divided into 3 experiment groups including vehicle group (SHR/Ve), low-dose telmisartan group (SHR/Low, .3 mg/kg/day), and high-dose telmisartan group (SHR/High, 3 mg/kg/day). RESULTS: The numbers of LDL-R- and immuno-ApoE-positive neurons increased in both cerebral cortex and hippocampus of SHR/Ve throughout 6, 12, and 18 months of age, compared with age-matched normotensive Wistar rats. On the other hand, telmisartan significantly reduced the numbers of LDL-R- and ApoE immuno-positive neurons in both cerebral cortex and hippocampus, with similar effectiveness in the SHR/Low group without blood pressure (BP) lowering to BP lowering (SHR/High). The decrease of MAP2-positive neuron in SHR/Ve was recovered by telmisartan in both cerebral cortex and hippocampus. CONCLUSIONS: These findings suggest that a long-term treatment with telmisartan directly improved neuronal lipid metabolism in the cerebral cortex and hippocampus of SHR-SR, mainly improving LDL-R and ApoE metabolism (SHR/Low) with a small additive benefit by BP lowering (SHR/High), which could provide a preventative approach in patients with hypertension at risk of Alzheimer disease.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Apolipoproteins E/biosynthesis , Apolipoproteins E/drug effects , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cerebral Cortex/metabolism , Hippocampus/metabolism , Receptors, LDL/biosynthesis , Receptors, LDL/drug effects , Stroke/genetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Dementia, Vascular/pathology , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/enzymology , Male , Mitogen-Activated Protein Kinase 1/biosynthesis , Rats , Rats, Inbred SHR , Rats, Wistar , Telmisartan
17.
Lipids Health Dis ; 13: 136, 2014 Aug 23.
Article in English | MEDLINE | ID: mdl-25148848

ABSTRACT

Excessive energy storage of adipose tissue makes contribution to the occurrence and progression of obesity, which accompanies with multiple adverse complications, such as metabolic syndrome, cardiovascular diseases. It is well known that apolipoprotein E, as a component of lipoproteins, performs a key role in maintaining plasma lipoproteins homeostasis. Interestingly, apolipoprotein E is highly expressed in adipocyte and has positive relation with body fat mass. Apolipoprotein E knock-out mice show small fat mass compared to wild type mice. Moreover, adipocyte deficiency in apolipoprotein E shows impaired lipoproeteins internalization and triglyceride accumulation. Apolipopreotein E-deficient lipoproteins can not induce preadipocyte to form round full-lipid adipocyte, whereas apolipoprotein E-containing lipoproteins can. This article mainly reviews the modulation of apolipoprotein E synthesized by adipocyte and apolipoprotein E carried on lipoproteins in adipocyte triglyceride content.


Subject(s)
Adipocytes/metabolism , Apolipoproteins E/biosynthesis , Triglycerides/metabolism , Animals , Humans , Lipoproteins, VLDL/metabolism , Obesity/metabolism
18.
J Gen Virol ; 95(Pt 7): 1510-1521, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24744301

ABSTRACT

Orphan nuclear receptor subfamily 4 group A member 1 (NR4A1) is a transcription factor stimulated by many factors and plays pivotal roles in metabolism, proliferation and apoptosis. In this study, the expression of NR4A1 in Huh7.5.1 cells was significantly upregulated by hepatitis C virus (HCV) infection. The silencing of NR4A1 inhibited the entry of HCV and reduced the specific infectivity of secreted HCV particles but had only minor or no effect on the genome replication and translation, virion assembly and virus release steps of the virus life cycle. Further experiments demonstrated that the silencing of NR4A1 affected virus entry through pan-downregulation of the expression of HCV receptors scavenger receptor BI, occludin, claudin-1 and epidermal growth factor receptor but not CD81. The reduced specific infectivity of HCV in the knockdown cells was due to decreased apolipoprotein E (ApoE) expression. These results explain the delayed spread of HCV in NR4A1 knockdown Huh7.5.1 cells. Thus, NR4A1 plays a role in HCV replication through regulating the expression of HCV receptors and ApoE, and facilitates HCV entry and spread.


Subject(s)
Apolipoproteins E/biosynthesis , Hepacivirus/physiology , Hepatocytes/virology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Virus/biosynthesis , Virus Internalization , Cell Line, Tumor , Gene Expression Regulation , Gene Silencing , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
19.
PLoS One ; 8(11): e79908, 2013.
Article in English | MEDLINE | ID: mdl-24244577

ABSTRACT

The apoE production by tissue macrophages is crucial for the prevention of atherosclerosis and the aim of this study was to further elucidate how this apolipoprotein is regulated by cytokines present during inflammation. Here we studied apoE production in peripheral blood mononuclear cells (PBMC) and analysis was made with a newly developed apoE ELISpot assay. In PBMC, apoE secretion was restricted to monocytes with classical (CD14(++)CD16(-)) and intermediate (CD14(+)CD16(+)) monocytes being the main producers. As earlier described for macrophages, production was strongly upregulated by TGF-ß and downregulated by bacterial lipopolysaccharide (LPS) and the inflammatory cytokines IFN-γ, TNF-α and IL-1ß. We could here show that a similar down-regulatory effect was also observed with the type I interferon, IFN-α, while IL-6, often regarded as one of the more prominent inflammatory cytokines, did not affect TGF-ß-induced apoE production. The TNF-α inhibitor Enbrel could partly block the down-regulatory effect of IFN-γ, IFN-α and IL-1ß, indicating that inhibition of apoE by these cytokines may be dependent on or synergize with TNF-α. Other cytokines tested, IL-2, IL-4, IL-12, IL-13, IL-17A and IL-23, had no inhibitory effect on apoE production. In contrast to the effect on monocytes, apoE production by primary hepatocytes and the hepatoma cell line HepG2 was more or less unaffected by treatment with cytokines or LPS.


Subject(s)
Apolipoproteins E/biosynthesis , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Monocytes/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Apolipoproteins E/immunology , Biomarkers/metabolism , Etanercept , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/immunology , Humans , Immunoglobulin G/pharmacology , Immunologic Factors/pharmacology , Interleukin-6/pharmacology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Monocytes/cytology , Monocytes/immunology , Primary Cell Culture , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, Tumor Necrosis Factor , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
20.
Biochem Biophys Res Commun ; 441(2): 321-6, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24140409

ABSTRACT

Cholesterol efflux from macrophages is a critical mechanism to prevent the development of atherosclerosis. Here, we sought to investigate the effects of arctigenin, a bioactive component of Arctium lappa, on the cholesterol efflux in oxidized low-density lipoprotein (oxLDL)-loaded THP-1 macrophages. Our data showed that arctigenin significantly accelerated apolipoprotein A-I- and high-density lipoprotein-induced cholesterol efflux in both dose- and time-dependent manners. Moreover, arctigenin treatment enhanced the expression of ATP binding cassette transporter A1 (ABCA1), ABCG1, and apoE, all of which are key molecules in the initial step of cholesterol efflux, at both mRNA and protein levels. Arctigenin also caused a concentration-dependent elevation in the expression of peroxisome proliferator-activated receptor-gamma (PPAR-γ) and liver X receptor-alpha (LXR-α). The arctigenin-mediated induction of ABCA1, ABCG1, and apoE was abolished by specific inhibition of PPAR-γ or LXR-α using small interfering RNA technology. Our results collectively indicate that arctigenin promotes cholesterol efflux in oxLDL-loaded THP-1 macrophages through upregulation of ABCA1, ABCG1 and apoE, which is dependent on the enhanced expression of PPAR-γ and LXR-α.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Furans/pharmacology , Lignans/pharmacology , Macrophages/drug effects , Orphan Nuclear Receptors/metabolism , PPAR gamma/metabolism , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/biosynthesis , Apolipoproteins E/biosynthesis , Cell Line , Humans , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...