Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Br J Pharmacol ; 181(17): 3118-3135, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38710660

RESUMO

BACKGROUND AND PURPOSE: Traumatic brain injury (TBI) remains a major public health concern worldwide with unmet effective treatment. Stimulator of interferon genes (STING) and its downstream type-I interferon (IFN) signalling are now appreciated to be involved in TBI pathogenesis. Compelling evidence have shown that STING and type-I IFNs are key in mediating the detrimental neuroinflammatory response after TBI. Therefore, pharmacological inhibition of STING presents a viable therapeutic opportunity in combating the detrimental neuroinflammatory response after TBI. EXPERIMENTAL APPROACH: This study investigated the neuroprotective effects of the small-molecule STING inhibitor n-(4-iodophenyl)-5-nitrofuran-2-carboxamide (C-176) in the controlled cortical impact mouse model of TBI in 10- to 12-week-old male mice. Thirty minutes post-controlled cortical impact surgery, a single 750-nmol dose of C-176 or saline (vehicle) was administered intravenously. Analysis was conducted 2 h and 24 h post-TBI. KEY RESULTS: Mice administered C-176 had significantly smaller cortical lesion area when compared to vehicle-treated mice 24 h post-TBI. Quantitative temporal gait analysis conducted using DigiGait™ showed C-176 administration attenuated TBI-induced impairments in gait symmetry, stride frequency and forelimb stance width. C-176-treated mice displayed a significant reduction in striatal gene expression of pro-inflammatory cytokines Tnf-α, Il-1ß and Cxcl10 compared to their vehicle-treated counterparts 2 h post-TBI. CONCLUSION AND IMPLICATIONS: This study demonstrates the neuroprotective activity of C-176 in ameliorating acute neuroinflammation and preventing white matter neurodegeneration post-TBI. This study highlights the therapeutic potential of small-molecule inhibitors targeting STING for the treatment of trauma-induced inflammation and neuroprotective potential.


Assuntos
Lesões Encefálicas Traumáticas , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias , Fármacos Neuroprotetores , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Masculino , Camundongos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/administração & dosagem , Modelos Animais de Doenças
2.
Mol Psychiatry ; 26(10): 5516-5531, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34400772

RESUMO

Amyloidogenic processing of the amyloid precursor protein (APP) forms the amyloid-ß peptide (Aß) component of pathognomonic extracellular plaques of AD. Additional early cortical changes in AD include neuroinflammation and elevated iron levels. Activation of the innate immune system in the brain is a neuroprotective response to infection; however, persistent neuroinflammation is linked to AD neuropathology by uncertain mechanisms. Non-parametric machine learning analysis on transcriptomic data from a large neuropathologically characterised patient cohort revealed the acute phase protein lactoferrin (Lf) as the key predictor of amyloid pathology. In vitro studies showed that an interaction between APP and the iron-bound form of Lf secreted from activated microglia diverted neuronal APP endocytosis from the canonical clathrin-dependent pathway to one requiring ADP ribosylation factor 6 trafficking. By rerouting APP recycling to the Rab11-positive compartment for amyloidogenic processing, Lf dramatically increased neuronal Aß production. Lf emerges as a novel pharmacological target for AD that not only modulates APP processing but provides a link between Aß production, neuroinflammation and iron dysregulation.


Assuntos
Doença de Alzheimer , Lactoferrina , Proteínas de Fase Aguda , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Humanos
3.
J Neurochem ; 156(6): 979-987, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32614980

RESUMO

Glutaminyl cyclases (QC) catalyze the formation of neurotoxic pGlu-modified amyloid-ß peptides found in the brains of people with Alzheimer's disease (AD). Reports of several-fold increases in soluble QC (sQC) expression in the brain and peripheral circulation of AD individuals has prompted the development of QC inhibitors as potential AD therapeutics. There is, however, a lack of standardized quantitative data on QC expression in human tissues, precluding inter-laboratory comparison and validation. We tested the hypothesis that QC is elevated in AD tissues by quantifying levels of sQC protein and activity in post-mortem brain tissues from AD and age-matched control individuals. We found a modest but statistically significant increase in sQC protein, which paralleled a similar increase in enzyme activity. In plasma samples sourced from the Australian Imaging, Biomarker and Lifestyle study we determined that QC activity was not different between the AD and control group, though a modest increase was observed in female AD individuals compared to controls. Plasma QC activity was further correlated with levels of circulating monocytes in AD individuals. These data provide quantitative evidence that alterations in QC expression are associated with AD pathology.


Assuntos
Doença de Alzheimer/enzimologia , Aminoaciltransferases/metabolismo , Encéfalo/enzimologia , Idoso , Idoso de 80 Anos ou mais , Aminoaciltransferases/antagonistas & inibidores , Aminoaciltransferases/sangue , Austrália , Autopsia , Biomarcadores , Bases de Dados Factuais , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Estilo de Vida , Masculino , Pessoa de Meia-Idade , Valores de Referência , Caracteres Sexuais
4.
Mol Psychiatry ; 25(9): 1958-1966, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32444869

RESUMO

The proteolytic cleavage of ß-amyloid precursor protein (APP) to form the amyloid beta (Aß) peptide is related to the pathogenesis of Alzheimer's disease (AD) because APP mutations that influence this processing either induce familial AD or mitigate the risk of AD. Yet Aß formation itself may not be pathogenic. APP promotes neuronal iron efflux by stabilizing the cell-surface presentation of ferroportin, the only iron export channel of cells. Mislocalization of APP can promote iron retention, thus we hypothesized that changes in endocytotic trafficking associated with altered APP processing could contribute to the neuronal iron elevation and oxidative burden that feature in AD pathology. Here, we demonstrate, using genetic and pharmacological approaches, that endocytotic amyloidogenic processing of APP impairs iron export by destabilizing ferroportin on the cell surface. Conversely, preferential non-amyloidogenic processing of APP at the cell surface promotes ferroportin stabilization to decrease intraneuronal iron. A new Aß-independent hypothesis emerges where the amyloidogenic processing of APP, combined with age-dependent iron elevation in the tissue, increases pro-oxidant iron burden in AD.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Humanos , Ferro , Neurônios
5.
Neurochem Res ; 44(6): 1367-1374, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30796750

RESUMO

Cell surface ß-Amyloid precursor protein (APP) is known to have a functional role in iron homeostasis through stabilising the iron export protein ferroportin (FPN). Mechanistic evidence of this role has previously only been provided through transcriptional or translational depletion of total APP levels. However, numerous post-translational modifications of APP are reported to regulate the location and trafficking of this protein to the cell surface. Stable overexpressing cell lines were generated that overexpressed APP with disrupted N-glycosylation (APPN467K and APPN496K) or ectodomain phosphorylation (APPS206A); sites selected for their proximity to the FPN binding site on the E2 domain of APP. We hypothesise that impaired N-glycosylation or phosphorylation of APP disrupts the functional location on the cell surface or binding to FPN to consequentially alter intracellular iron levels through impaired cell surface FPN stability. Outcomes confirm that these post-translational modifications are essential for the correct location of APP on the cell surface and highlight a novel mechanism by which the cell can modulate iron homeostasis. Further interrogation of other post-translational processes to APP is warranted in order to fully understand how each modification plays a role on regulating intracellular iron levels in health and disease.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Homeostase/fisiologia , Ferro/metabolismo , Neurônios/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Glicosilação , Camundongos , Fosforilação/genética , Mutação Puntual , Processamento de Proteína Pós-Traducional/genética , Transporte Proteico/genética
6.
Neurotherapeutics ; 15(4): 1055-1062, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30112699

RESUMO

Proteolytic cleavage of the amyloid precursor protein (APP) into the Aß peptide has been an extensively researched mechanism for Alzheimer's disease, but the normal function of the protein is less understood. APP functions to regulate neuronal iron content by stabilizing the surface presentation of ferroportin-the only iron exporter channel of cells. The present study aims to quantify the contribution of APP to brain and peripheral iron by examining the lifetime impact on brain and liver iron levels in APP knockout mice. Consistent with previous reports, we found that wild-type mice exhibited an age-dependent increase in iron and ferritin in the brain, while no age-dependent changes were observed in the liver. APP ablation resulted in an exaggeration of age-dependent iron accumulation in the brain and liver in mice that was assessed at 8, 12, 18, and 22 months of age. Brain ferroportin levels were decreased in APP knockout mice, consistent with a mechanistic role for APP in stabilizing this iron export protein in the brain. Iron elevation in the brain and liver of APP knockout mice correlated with decreased transferrin receptor 1 and increased ferritin protein levels. However, no age-dependent increase in brain ferritin iron saturation was observed in APP-KO mice despite similar protein expression levels potentially explaining the vulnerability of APP-KO mice to parkinsonism and traumatic brain sequelae. Our results support a crucial role of APP in regulating brain and peripheral iron, and show that APP may act to oppose brain iron elevation during aging.


Assuntos
Envelhecimento/patologia , Precursor de Proteína beta-Amiloide/deficiência , Encéfalo/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Fatores Etários , Precursor de Proteína beta-Amiloide/genética , Animais , Ferritinas/metabolismo , Camundongos , Camundongos Knockout
7.
Sci Rep ; 7(1): 9835, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852095

RESUMO

Amyloid precursor protein (APP) and its extracellular domain, soluble APP alpha (sAPPα) play important physiological and neuroprotective roles. However, rare forms of familial Alzheimer's disease are associated with mutations in APP that increase toxic amyloidogenic cleavage of APP and produce amyloid beta (Aß) at the expense of sAPPα and other non-amyloidogenic fragments. Although mitochondrial dysfunction has become an established hallmark of neurotoxicity, the link between Aß and mitochondrial function is unclear. In this study we investigated the effects of increased levels of neuronal APP or Aß on mitochondrial metabolism and gene expression, in human SH-SY5Y neuroblastoma cells. Increased non-amyloidogenic processing of APP, but not Aß, profoundly decreased respiration and enhanced glycolysis, while mitochondrial DNA (mtDNA) transcripts were decreased, without detrimental effects to cell growth. These effects cannot be ascribed to Aß toxicity, since higher levels of endogenous Aß in our models do not cause oxidative phosphorylation (OXPHOS) perturbations. Similarly, chemical inhibition of ß-secretase decreased mitochondrial respiration, suggesting that non-amyloidogenic processing of APP may be responsible for mitochondrial changes. Our results have two important implications, the need for caution in the interpretation of mitochondrial perturbations in models where APP is overexpressed, and a potential role of sAPPα or other non-amyloid APP fragments as acute modulators of mitochondrial metabolism.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular , Respiração Celular/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ativação Enzimática , Dosagem de Genes , Genes Mitocondriais , Glicólise , Humanos , Mitocôndrias/genética , Mutação , Neurônios/metabolismo , Transcrição Gênica
8.
Mol Neurodegener ; 12(1): 45, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592304

RESUMO

Parkinson's disease is a multifactorial neurodegenerative disorder, the aetiology of which remains elusive. The primary clinical feature of progressively impaired motor control is caused by a loss of midbrain substantia nigra dopamine neurons that have a high α-synuclein (α-syn) and iron content. α-Syn is a neuronal protein that is highly modified post-translationally and central to the Lewy body neuropathology of the disease. This review provides an overview of findings on the role post translational modifications to α-syn have in membrane binding and intracellular vesicle trafficking. Furthermore, we propose a concept in which acetylation and phosphorylation of α-syn modulate endocytic import of iron and vesicle transport of dopamine during normal physiology. Disregulated phosphorylation and oxidation of α-syn mediate iron and dopamine dependent oxidative stress through impaired cellular location and increase propensity for α-syn aggregation. The proposition highlights a connection between α-syn, iron and dopamine, three pathological components associated with disease progression in sporadic Parkinson's disease.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Ferro/metabolismo , Doença de Parkinson/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , alfa-Sinucleína/metabolismo , Animais , Dopamina/metabolismo , Humanos
9.
ACS Chem Neurosci ; 8(4): 731-736, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28029772

RESUMO

Ferroxidase activity has been reported to be altered in various biological fluids in neurodegenerative disease, but the sources contributing to the altered activity are uncertain. Here we assay fractions of serum and cerebrospinal fluid with a newly validated triplex ferroxidase assay. Our data indicate that while ceruloplasmin, a multicopper ferroxidase, is the predominant source of serum activity, activity in CSF predominantly derives from a <10 kDa component, specifically from polyanions such as citrate and phosphate. We confirm that in human biological samples, ceruloplasmin activity in serum is decreased in Alzheimer's disease, but in CSF a reduction of activity in Alzheimer's disease originates from the polyanion component.


Assuntos
Doença de Alzheimer/metabolismo , Líquido Cefalorraquidiano/enzimologia , Ceruloplasmina/metabolismo , Ferro/metabolismo , Soro/enzimologia , Ceruloplasmina/análise , Humanos , Oxirredução
10.
J Biol Chem ; 291(12): 6134-45, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26697885

RESUMO

Pyroglutamate-modified amyloid-ß (pE-Aß) is a highly neurotoxic amyloid-ß (Aß) isoform and is enriched in the brains of individuals with Alzheimer disease compared with healthy aged controls. Pyroglutamate formation increases the rate of Aß oligomerization and alters the interactions of Aß with Cu(2+) and lipids; however, a link between these properties and the toxicity of pE-Aß peptides has not been established. We report here that Aß3pE-42 has an enhanced capacity to cause lipid peroxidation in primary cortical mouse neurons compared with the full-length isoform (Aß(1-42)). In contrast, Aß(1-42) caused a significant elevation in cytosolic reactive oxygen species, whereas Aß3pE-42 did not. We also report that Aß3pE-42 preferentially associates with neuronal membranes and triggers Ca(2+) influx that can be partially blocked by the N-methyl-d-aspartate receptor antagonist MK-801. Aß3pE-42 further caused a loss of plasma membrane integrity and remained bound to neurons at significantly higher levels than Aß(1-42) over extended incubations. Pyroglutamate formation was additionally found to increase the relative efficiency of Aß-dityrosine oligomer formation mediated by copper-redox cycling.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Sinalização do Cálcio , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Ascórbico/química , Permeabilidade da Membrana Celular , Células Cultivadas , Cobre/química , Humanos , Peroxidação de Lipídeos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos , Ácido Pirrolidonocarboxílico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
11.
PLoS One ; 9(12): e114174, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25464026

RESUMO

Ceruloplasmin is a ferroxidase that interacts with ferroportin to export cellular iron, but is not expressed in neurons. We recently reported that the amyloid precursor protein (APP) is the analogous iron-exporting chaperone for neurons and other cells. The ferroxidase activity of APP has since been called into question. Using a triplex Fe2+ oxidation assay, we analyzed the activity of a soluble form of APP (sAPPα) within a buffer of physiological pH and anionic charge, and determined that iron oxidation originated from phosphate. Using various techniques such as flow-cytometry to measure surface presented proteins, we confirmed that endogenous APP is essential for ferroportin persistence on the neuronal surface. Therefore, despite lacking ferroxidase activity, APP still supports iron export from neurons.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Ceruloplasmina/metabolismo , Animais , Células HEK293 , Humanos , Camundongos , Oxirredução
12.
Biochim Biophys Acta ; 1840(12): 3299-310, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25152357

RESUMO

BACKGROUND: Iron oxidation is thought to be predominantly handled enzymatically in the body, to minimize spontaneous combustion with oxygen and to facilitate cellular iron export by loading transferrin. This process may be impaired in disease, and requires more accurate analytical assays to interrogate enzymatic- and auto-oxidation within a physiologically relevant environment. METHOD: A new triplex ferroxidase activity assay has been developed that overcomes the previous assay limitations of measuring iron oxidation at a physiologically relevant pH and salinity. RESULTS: Revised enzymatic kinetics for ceruloplasmin (Vmax≈35µMFe(3+)/min/µM; Km≈15µM) are provided under physiological conditions, and inhibition by sodium azide (Ki for Ferric Gain 78.3µM, Ki for transferrin loading 8.1×10(4)µM) is quantified. We also used this assay to characterize the non-enzymatic oxidation of iron that proceeded linearly under physiological conditions. CONCLUSIONS AND GENERAL SIGNIFICANCE: These findings indicate that the requirement of an enzyme to oxidize iron may only be necessary under conditions of adverse pH or anionic strength, for example from hypoxia. In a normal physiological environment, Fe(3+) incorporation into transferrin would be sufficiently enabled by the biological polyanions that are prevalent within extracellular fluids.

13.
Front Aging Neurosci ; 6: 91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24860500

RESUMO

Alzheimer's disease (AD) is a multifactorial neurodegenerative disease. It begins years prior to the onset of clinical symptoms, such as memory loss and cognitive decline. Pathological hallmarks of AD include the accumulation of ß-amyloid in plaques and hyperphosphorylated tau in neurofibrillary tangles. Copper, iron, and zinc are abnormally accumulated and distributed in the aging brain. These metal ions can adversely contribute to the progression of AD. Dysregulation of cholesterol metabolism has also been implicated in the development of AD pathology. To date, large bodies of research have been carried out independently to elucidate the role of metals or cholesterol on AD pathology. Interestingly, metals and cholesterol affect parallel molecular and biochemical pathways involved in AD pathology. The possible links between metal dyshomeostasis and altered brain cholesterol metabolism in AD are reviewed.

14.
Front Pharmacol ; 5: 81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24795635

RESUMO

As with most bioavailable transition metals, iron is essential for many metabolic processes required by the cell but when left unregulated is implicated as a potent source of reactive oxygen species. It is uncertain whether the brain's evident vulnerability to reactive species-induced oxidative stress is caused by a reduced capability in cellular response or an increased metabolic activity. Either way, dys-regulated iron levels appear to be involved in oxidative stress provoked neurodegeneration. As in peripheral iron management, cells within the central nervous system tightly regulate iron homeostasis via responsive expression of select proteins required for iron flux, transport and storage. Recently proteins directly implicated in the most prevalent neurodegenerative diseases, such as amyloid-ß precursor protein, tau, α-synuclein, prion protein and huntingtin, have been connected to neuronal iron homeostatic control. This suggests that disrupted expression, processing, or location of these proteins may result in a failure of their cellular iron homeostatic roles and augment the common underlying susceptibility to neuronal oxidative damage that is triggered in neurodegenerative disease.

15.
Ann Neurol ; 73(4): 554-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23424051

RESUMO

Ceruloplasmin is an iron-export ferroxidase that is abundant in plasma and also expressed in glia. We found a ∼80% loss of ceruloplasmin ferroxidase activity in the substantia nigra of idiopathic Parkinson disease (PD) cases, which could contribute to the pro-oxidant iron accumulation that characterizes the pathology. Consistent with a role for ceruloplasmin in PD etiopathogenesis, ceruloplasmin knockout mice developed parkinsonism that was rescued by iron chelation. Additionally, peripheral infusion of ceruloplasmin attenuated neurodegeneration and nigral iron elevation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model for PD. These findings show, in principle, that intravenous ceruloplasmin may have therapeutic potential in PD.


Assuntos
Ceruloplasmina/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Idoso , Animais , Estudos de Casos e Controles , Ceruloplasmina/deficiência , Ceruloplasmina/uso terapêutico , Deferiprona , Modelos Animais de Doenças , Dopaminérgicos , Feminino , Humanos , Quelantes de Ferro/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Doença de Parkinson/etiologia , Doença de Parkinson/patologia , Piridonas/uso terapêutico , Substância Negra/metabolismo
16.
Nat Med ; 18(2): 291-5, 2012 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-22286308

RESUMO

The microtubule-associated protein tau has risk alleles for both Alzheimer's disease and Parkinson's disease and mutations that cause brain degenerative diseases termed tauopathies. Aggregated tau forms neurofibrillary tangles in these pathologies, but little is certain about the function of tau or its mode of involvement in pathogenesis. Neuronal iron accumulation has been observed pathologically in the cortex in Alzheimer's disease, the substantia nigra (SN) in Parkinson's disease and various brain regions in the tauopathies. Here we report that tau-knockout mice develop age-dependent brain atrophy, iron accumulation and SN neuronal loss, with concomitant cognitive deficits and parkinsonism. These changes are prevented by oral treatment with a moderate iron chelator, clioquinol. Amyloid precursor protein (APP) ferroxidase activity couples with surface ferroportin to export iron, but its activity is inhibited in Alzheimer's disease, thereby causing neuronal iron accumulation. In primary neuronal culture, we found loss of tau also causes iron retention, by decreasing surface trafficking of APP. Soluble tau levels fall in affected brain regions in Alzheimer's disease and tauopathies, and we found a similar decrease of soluble tau in the SN in both Parkinson's disease and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. These data suggest that the loss of soluble tau could contribute to toxic neuronal iron accumulation in Alzheimer's disease, Parkinson's disease and tauopathies, and that it can be rescued pharmacologically.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Demência/etiologia , Ferro/metabolismo , Transtornos Parkinsonianos/etiologia , Proteínas tau/deficiência , Fatores Etários , Idoso , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Encéfalo/metabolismo , Química Encefálica , Ceruloplasmina/metabolismo , Ceruloplasmina/fisiologia , Demência/metabolismo , Humanos , Ferro/análise , Masculino , Camundongos , Camundongos Knockout , Transtornos Parkinsonianos/metabolismo
17.
Lipids ; 46(10): 931-41, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21674150

RESUMO

Type 2 diabetes and dyslipidemia are risk factors for cardiovascular disease. However, mechanisms by which hypertriglyceridemia influences atherogenesis remain unclear. We examined effects of dyslipidemic diabetic serum on macrophage lipid accumulation as a model of foam cell formation. Normal human macrophages were cultured in media supplemented with 10% serum from non-diabetic normolipidemic or non-diabetic hypercholesterolemic adults versus adults with Type 2 diabetes; diabetes and hypertriglyceridemia; or diabetes and hypercholesterolemia. Exposure to diabetic sera resulted in increased macrophage fatty acids (2-3 fold higher, both saturated and unsaturated). Macrophage expression of CD36, scavenger receptor A (SR-A) and stearoyl-CoA desaturase (SCD) was increased, most prominently in macrophages exposed to hypertriglyceridemic diabetic serum (twofold increase in CD36 and fourfold increase in SCD, p < 0.05). In these conditions, RNA inhibition of CD36 reduced macrophage free cholesterol (163.9 ± 10.5 vs. 221.9 ± 26.2 mmol free cholesterol/g protein, p = 0.04). RNA inhibition of SCD decreased macrophage fatty acid content, increased ABCA1 level and enhanced cholesterol efflux (18.0 ± 3.9 vs. 8.0 ± 0.8% at 48 h, p = 0.03). Diabetic dyslipidemia may contribute to accelerated atherosclerosis via alterations in macrophage lipid metabolism favoring foam cell formation. Increased expression of CD36 and SR-A would facilitate macrophage lipid uptake, while increased expression of SCD could block compensatory upregulation of ABCA1 and cholesterol efflux. Further studies are needed to clarify whether modulation of macrophage lipid metabolism might reduce progression of diabetic atherosclerosis.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Dislipidemias/sangue , Metabolismo dos Lipídeos , Macrófagos/enzimologia , Estearoil-CoA Dessaturase/metabolismo , Adulto , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Dislipidemias/complicações , Ácidos Graxos/metabolismo , Células Espumosas/metabolismo , Humanos , RNA Interferente Pequeno/genética , Receptores Depuradores Classe A/metabolismo , Estearoil-CoA Dessaturase/genética
18.
Lipids ; 46(4): 371-80, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21286835

RESUMO

Macrophages in arterial walls accumulate lipids leading to the development of atherosclerotic plaques. However, mechanisms underlying macrophage lipid accumulation and foam cell formation are often studied without accounting for risk factors such as dyslipidemia. We investigated the effect of varying concentrations of triglyceride (TG) within physiological range on macrophage fatty acid (FA) accumulation and expression of cholesterol efflux proteins. Human monocytes were cultured in media supplemented with 10% sera containing low (0.7 mmol/L) to high (1.4 mmol/L) TG. The resulting macrophages were harvested after 10 days for analysis of FA content and composition and expression of genes involved in lipid metabolism. Exposure to higher TG and lower HDL concentrations in media increased macrophage lipid content. Macrophages exposed to higher TG had increased total FA content compared with controls (876 µg/mg protein vs. 652 µg/mg protein) and greater proportions of C16:0, C18:1 and C18:2. Macrophage expression of both ABCA1 and ABCG1 cholesterol efflux proteins were reduced when higher TG concentrations were present in the media. Expression of scavenger receptor CD36, involved in lipoprotein uptake, was also downregulated in macrophages exposed to higher TG. Culturing macrophages in conditions of higher versus lower TG influenced macrophage FA content and composition, and levels of regulatory proteins. Replicating in vitro levels of dyslipidemia encountered in vivo may provide an informative model for investigation of atherogenesis.


Assuntos
Dislipidemias/sangue , Ácidos Graxos/metabolismo , Macrófagos/química , Macrófagos/metabolismo , Soro/química , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Meios de Cultura/química , Ácidos Graxos/química , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Triglicerídeos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA