171 research outputs found

    High Saturated Fat-Enriched Diet Evokes Endoplasmic Reticulum Stress and Consequently Increases ?- Site App Cleaving Enzyme 1 Activity in Amyloid-Beta Engenderment in the Brain

    Get PDF
    Alzheimer’s disease (AD) is the most common form of dementia in the elderly that is histo-pathologically characterized by extracellular accumulation of aggregated Amyloid-? (A?) peptide as neuritic senile plaques and the intracellular accumulation of aggregated hyperphosphorylated protein tau (?) as neurofibrillary tangles. The aspartyl protease BACE1 is indispensable for the engenderment of A? and catalyzes the rate-limiting step in A? genesis from A?PP. The expression of BACE1 protein as well as its enzymatic activity is significantly augmented in the AD brain. The etiology of AD is multifactorial and egregiously comprehended, but epidemiological studies have implicated a diet rich in saturated free fatty acids (sFFA) as a significant risk factor for developing AD. Palmitic acid (palmitate) is the most abundant long-chain free saturated fatty acid in the brain and the diet and higher palmitate levels in the plasma, as observed in obesity and diabetes, inversely correlate with cognitive function. Recent cogent evidence has implicated endoplasmic reticulum (ER) stress as one of the culpable factors in initiating and fostering the deleterious neurodegenerative changes in AD. A multitude of studies have cogently demonstrated that sFFA such as palmitic acid evoke ER stress. In this study we demonstrate that palmitate evokes ER stress leading to the induction of CHOP expression which indispensably mediates the up-regulation in BACE1 expression and A? engenderment via the NF-?B signaling pathway. Our study unveils a novel ER stress/CHOP/NF-?B signaling pathway and delineates the molecular mechanism thereof that mediate the palmitate-induced up-regulation of BACE1 expression

    Endoplasmic Renticulum-Activated C/EBP Homologous Protein Mediates the Palmitate-Enriched Diet Induced Increase in the Lipogenic Expression in the Liver

    Get PDF
    Non-alcoholic fatty liver disease (NAFLD) is a wide spectrum pathophysiological disorder characterized by insulin resistance, hepatic steatosis, and inflammation. Diets rich in saturated fat are known to evoke insulin resistance, ER stress, and de novo lipogenesis and thereby contribute to the pathogenic mechanisms involved in NAFLD. Palmitic acid (palmitate) is the most abundant saturated fatty acid in the diet and palmitate-enriched diets are known to cause NAFLD in a multitude of rodent models of NAFLD. Palmitate-enriched diets are known to induce steatosis by inducing the expression of genes involved in de novo lipogenesis. However, the signaling mechanisms and the downstream molecular mediators involved have not been elucidated. In this study, we explored the role of palmitate-induced ER stress and subsequent induction of C/EBP Homologous Protein (CHOP) expression in the modulation of expression and transcriptional activities of Liver X Receptor alpha (LXR?) and Sterol Response Element Binding Protein 1c (SREBP1c), two indispensable transcription factors and master regulators of genes involved in de novo lipogenesis. We demonstrate, in exogenous palmitate-treated HepG2 cells and in the livers of palmitate-enriched diet-fed mice, that palmitate evokes ER stress leading to the induction of CHOP expression. We further show that CHOP mediates the up-regulation in expression levels and transcriptional activities of LXR? and SREBP1c. Our study identifies a unique ER stress-CHOP-LXR?/SREBP1c signaling pathway that mediates palmitate-induced up-regulation of lipogenic gene expression in the liver that may play a critical role in the etiopathogenesis of NAFLD

    Molecular interplay between leptin, insulin-like growth factor-1, and β-amyloid in organotypic slices from rabbit hippocampus

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Evidence shows that the insulin-like growth factor-1 (IGF-1) and leptin reduce β-amyloid (Aβ) production and tau phosphorylation, two major hallmarks of Alzheimer's disease (AD). IGF-1 expression involves the JAK/STAT pathway and the expression of leptin is regulated by the mammalian target of rapamycin complex 1 (mTORC1). We have previously shown that Aβ reduces leptin by inhibiting the mTORC1 pathway and Aβ was also suggested to inhibit the JAK/STAT pathway, potentially attenuating IGF-1 expression. As IGF-1 can activate mTORC1 and leptin can modulate JAK/STAT pathway, we determined the extent to which IGF-1 and leptin can upregulate the expression of one another and protect against Aβ-induced downregulation.</p> <p>Results</p> <p>We demonstrate that incubation of organotypic slices from adult rabbit hippocampus with Aβ42 downregulates IGF-1 expression by inhibiting JAK2/STAT5 pathway. Leptin treatment reverses these Aβ42 effects on IGF-1 and treatment with the STAT5 inhibitor completely abrogated the leptin-induced increase in IGF-1. Furthermore, EMSA and ChIP analyses revealed that leptin increases the STAT5 binding to the IGF-1 promoter. We also show that IGF-1 increases the expression of leptin and reverses the Aβ42-induced attenuation in leptin expression via the activation of mTORC1 signaling as the mTORC1 inhibitor rapamycin completely precluded the IGF-1-induced increase in leptin expression.</p> <p>Conclusion</p> <p>Our results demonstrate for the first time that Aβ42 downregulates IGF-1 expression and that leptin and IGF-1 rescue one another from downregulation by Aβ42. Our study provides a valuable insight into the leptin/IGF-1/Aβ interplay that may be relevant to the pathophysiology of AD.</p

    Cholesterol-enriched diet causes age-related macular degeneration-like pathology in rabbit retina

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Alzheimer's disease (AD) and age-related macular degeneration (AMD) share several pathological hallmarks including β-amyloid (Aβ) accumulation, oxidative stress, and apoptotic cell death. The causes of AD and AMD are likely multi-factorial with several factors such as diet, environment, and genetic susceptibility participating in the pathogenesis of these diseases. Epidemiological studies correlated high plasma cholesterol levels with high incidence of AD, and feeding rabbits with a diet rich in cholesterol has been shown to induce AD-like pathology in rabbit brain. High intake of cholesterol and saturated fat were also long been suspected to increase the risk for AMD. However, the extent to which cholesterol-enriched diet may also cause AMD-like features in rabbit retinas is not well known.</p> <p>Methods</p> <p>Male New Zealand white rabbits were fed normal chow or a 2% cholesterol-enriched diet for 12 weeks. At necropsy, animals were perfused with Dulbecco's phosphate-buffered saline and the eyes were promptly removed. One eye of each animal was used for immunohistochemistry and retina dissected from the other eye was used for Western blot, ELISA assays, spectrophotometry and mass spectrometry analyses.</p> <p>Results</p> <p>Increased levels of Aβ, decreased levels of the anti-apoptotic protein Bcl-2, increased levels of the pro-apoptotic Bax and gadd153 proteins, emergence of TUNEL-positive cells, and increased generation of reactive oxygen species were found in retinas from cholesterol-fed compared to normal chow-fed rabbits. Additionally, astrogliosis, drusen-like debris and cholesterol accumulations in retinas from cholesterol-fed rabbits were observed. As several lines of evidence suggest that oxidized cholesterol metabolites (oxysterols) may be the link by which cholesterol contributes to the pathogenesis of AMD, we determined levels of oxysterols and found a dramatic increase in levels of oxysterols in retinas from cholesterol-fed rabbits.</p> <p>Conclusions</p> <p>Our results suggest that cholesterol-enriched diets cause retinal degeneration that is relevant to AMD. Furthermore, our data suggests high cholesterol levels and subsequent increase in the cholesterol metabolites as potential culprits to AMD.</p

    Oxidised LDL-lipids increase beta amyloid production by SH-SY5Y cells through glutathione depletion and lipid raft formation

    Get PDF
    Elevated total cholesterol in midlife has been associated with increased risk of dementia in later life. We have previously shown that low-density lipoprotein (LDL) is more oxidized in the plasma of dementia patients, although total cholesterol levels are not different from those of age-matched controls. β-Amyloid (Aβ) peptide, which accumulates in Alzheimer disease (AD), arises from the initial cleavage of amyloid precursor protein by β-secretase-1 (BACE1). BACE1 activity is regulated by membrane lipids and raft formation. Given the evidence for altered lipid metabolism in AD, we have investigated a mechanism for enhanced Aβ production by SH-SY5Y neuronal-like cells exposed to oxidized LDL (oxLDL). The viability of SH-SY5Y cells exposed to 4 μg oxLDL and 25 μM 27-hydroxycholesterol (27OH-C) was decreased significantly. Lipids, but not proteins, extracted from oxLDL were more cytotoxic than oxLDL. In parallel, the ratio of reduced glutathione (GSH) to oxidized glutathione was decreased at sublethal concentrations of lipids extracted from native and oxLDL. GSH loss was associated with an increase in acid sphingomyelinase (ASMase) activity and lipid raft formation, which could be inhibited by the ASMase inhibitor desipramine. 27OH-C and total lipids from LDL and oxLDL independently increased Aβ production by SH-SY5Y cells, and Aβ accumulation could be inhibited by desipramine and by N-acetylcysteine. These data suggest a mechanism whereby oxLDL lipids and 27OH-C can drive Aβ production by GSH depletion, ASMase-driven membrane remodeling, and BACE1 activation in neuronal cells. © 2014 The Authors

    Pomegranate inhibits neuroinflammation and amyloidogenesis in IL-1β stimulated SK-N-SH cells

    Get PDF
    Purpose: Pomegranate fruit, Punica granatum L. (Punicaceae) and its constituents have been shown to inhibit inflammation. In this study we aimed to assess the effects of freeze-dried pomegranate (PWE) on PGE2 production in IL-1β stimulated SK-N-SH cells. Methods: An enzyme immuno assay (EIA) was used to measure prostaglandin E2 (PGE2) production from supernatants of IL-1β stimulated SK-N-SH cells. Expression of COX-2, phospho-IκB and phospho-IKK proteins were evaluated, while NF-κB reporter gene assay was carried out in TNFα-stimulated HEK293 cells to determine the effect of PWE on NF-κB transactivation. Levels of BACE-1 and Aβ in SK-N-SH cells stimulated with IL-1β were measured with an in cell ELISA. Results: PWE (25-200 µg/ml) dose dependently reduced COX-2 dependent PGE2 production in SK-N-SH cells stimulated with IL-1β. Phosphorylation of IκB and IKK were significantly (p<0.001) inhibited by PWE (50- 200 µg/ml). Our studies also show that PWE (50-200 µg/ml) significantly (p<0.01) inhibited NF-κB transactivation in TNFα-stimulated HEK293 cells. Furthermore PWE inhibited BACE-1 and Aβ expression in SK-N-SH cells treated with IL-1β. Conclusions: Taken together, our study demonstrates that pomegranate inhibits inflammation, as well as amyloidogenesis in IL-1β-stimulated SK-N-SH cells. We propose that pomegranate is a potential nutritional strategy in slowing the progression of neurodegenerative disorders like Alzheimer’s disease

    The oxysterol 27-hydroxycholesterol increases β-amyloid and oxidative stress in retinal pigment epithelial cells

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Alzheimer's disease (AD) and age-related macular degeneration (AMD) share several pathological features including β-amyloid (Aβ) peptide accumulation, oxidative damage, and cell death. The causes of AD and AMD are not known but several studies suggest disturbances in cholesterol metabolism as a culprit of these diseases. We have recently shown that the cholesterol oxidation metabolite 27-hydroxycholesterol (27-OHC) causes AD-like pathology in human neuroblastoma SH-SY5Y cells and in organotypic hippocampal slices. However, the extent to which and the mechanisms by which 27-OHC may also cause pathological hallmarks related to AMD are ill-defined. In this study, the effects of 27-OHC on AMD-related pathology were determined in ARPE-19 cells. These cells have structural and functional properties relevant to retinal pigmented epithelial cells, a target in the course of AMD.</p> <p>Methods</p> <p>ARPE-19 cells were treated with 0, 10 or 25 μM 27-OHC for 24 hours. Levels of Aβ peptide, mitochondrial and endoplasmic reticulum (ER) stress markers, Ca<sup>2+ </sup>homeostasis, glutathione depletion, reactive oxygen species (ROS) generation, inflammation and cell death were assessed using ELISA, Western blot, immunocytochemistry, and specific assays.</p> <p>Results</p> <p>27-OHC dose-dependently increased Aβ peptide production, increased levels of ER stress specific markers caspase 12 and gadd153 (also called CHOP), reduced mitochondrial membrane potential, triggered Ca<sup>2+ </sup>dyshomeostasis, increased levels of the nuclear factor κB (NFκB) and heme-oxygenase 1 (HO-1), two proteins activated by oxidative stress. Additionally, 27-OHC caused glutathione depletion, ROS generation, inflammation and apoptotic-mediated cell death.</p> <p>Conclusions</p> <p>The cholesterol metabolite 27-OHC is toxic to RPE cells. The deleterious effects of this oxysterol ranged from Aβ accumulation to oxidative cell damage. Our results suggest that high levels of 27-OHC may represent a common pathogenic factor for both AMD and AD.</p

    Adipocytokines and CD34+ Progenitor Cells in Alzheimer's Disease

    Get PDF
    BACKGROUND: Alzheimer's disease (AD) and atherosclerosis share common vascular risk factors such as arterial hypertension and hypercholesterolemia. Adipocytokines and CD34(+) progenitor cells are associated with the progression and prognosis of atherosclerotic diseases. Their role in AD is not adequately elucidated. METHODS AND FINDINGS: In the present study, we measured in 41 patients with early AD and 37 age- and weight-matched healthy controls blood concentrations of adiponectin and leptin by enzyme linked immunoabsorbent assay and of CD34(+) progenitor cells using flow cytometry. We found significantly lower plasma levels of leptin in AD patients compared with the controls, whereas plasma levels of adiponectin did not show any significant differences (AD vs. control (mean ± SD): leptin:8.9 ± 5.6 ng/mL vs.16.3 ± 15.5 ng/mL;P = 0.038; adiponectin:18.5 ± 18.1 µg/mL vs.16.7 ± 8.9 µg/mL;P = 0.641). In contrast, circulating CD34(+) cells were significantly upregulated in AD patients (mean absolute cell count ± SD:253 ± 51 vs. 203 ± 37; P = 0.02) and showed an inverse correlation with plasma levels of leptin (r =  -0.248; P = 0.037). In logistic regression analysis, decreased leptin concentration (P = 0.021) and increased number of CD34(+) cells (P = 0.036) were both significantly associated with the presence of AD. According to multifactorial analysis of covariance, leptin serum levels were a significant independent predictor for the number of CD34(+) cells (P = 0.002). CONCLUSIONS: Our findings suggest that low plasma levels of leptin and increased numbers of CD34(+) progenitor cells are both associated with AD. In addition, the results of our study provide first evidence that increased leptin plasma levels are associated with a reduced number of CD34(+) progenitor cells in AD patients. These findings point towards a combined involvement of leptin and CD34(+) progenitor cells in the pathogenesis of AD. Thus, plasma levels of leptin and circulating CD34(+) progenitor cells could represent an important molecular link between atherosclerotic diseases and AD. Further studies should clarify the pathophysiological role of both adipocytokines and progenitor cells in AD and possible diagnostic and therapeutic applications
    corecore