Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurophysiol ; 132(1): 23-33, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38748407

RESUMO

The apolipoprotein E (APOE) gene has been studied due to its influence on Alzheimer's disease (AD) development and work in an APOE mouse model recently demonstrated impaired respiratory motor plasticity following spinal cord injury (SCI). Individuals with AD often copresent with obstructive sleep apnea (OSA) characterized by cessations in breathing during sleep. Despite the prominence of APOE genotype and sex as factors in AD progression, little is known about the impact of these variables on respiratory control. Ventilation is tightly regulated across many systems, with respiratory rhythm formation occurring in the brainstem but modulated in response to chemoreception. Alterations within these modulatory systems may result in disruptions of appropriate respiratory control and ultimately, disease. Using mice expressing two different humanized APOE alleles, we characterized how sex and the presence of APOE3 or APOE4 influences ventilation during baseline breathing (normoxia) and during respiratory challenges. We show that sex and APOE genotype influence breathing during hypoxic challenge, which may have clinical implications in the context of AD and OSA. In addition, female mice, while responding robustly to hypoxia, were unable to recover to baseline respiratory levels, emphasizing sex differences in disordered breathing.NEW & NOTEWORTHY This study is the first to use whole body plethysmography (WBP) to measure the impact of APOE alleles on breathing under normoxia and during adverse respiratory challenges in a targeted replacement Alzheimer's model. Both sex and genotype were shown to affect breathing under normoxia, hypoxic challenge, and hypoxic-hypercapnic challenge. This work has important implications regarding the impact of genetics on respiratory control as well as applications pertaining to conditions of disordered breathing including sleep apnea and neurotrauma.


Assuntos
Hipóxia , Animais , Feminino , Masculino , Hipóxia/fisiopatologia , Camundongos , Apolipoproteína E4/genética , Genótipo , Hipercapnia/fisiopatologia , Camundongos Transgênicos , Caracteres Sexuais , Respiração , Apolipoproteína E3/genética , Apolipoproteínas E/genética , Fatores Sexuais , Camundongos Endogâmicos C57BL
2.
Infect Immun ; 91(9): e0025123, 2023 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-37594272

RESUMO

Apolipoprotein E (ApoE) is a lipid transport protein that is hypothesized to suppress proinflammatory cytokine production, particularly after stimulation with Toll-like receptor (TLR) ligands such as lipopolysaccharide (LPS). Studies using transgenic ApoE human replacement mice (APOE) expressing one of three different allelic variants suggest that there is a hierarchy in terms of responsiveness to proinflammatory stimuli such as APOE4/E4 > APOE3/E3 > APOE2/E2. In this study, we test the hypothesis that APOE genotype can also predict susceptibility to infection with the facultative intracellular gram-positive bacterium Listeria monocytogenes. We found that bone-marrow-derived macrophages isolated from aged APOE4/E4 mice expressed elevated levels of nitric oxide synthase 2 and were highly resistant to in vitro infection with L. monocytogenes compared to APOE3/E3 and APOE2/E2 mice. However, we did not find statistically significant differences in cytokine or chemokine output from either macrophages or whole splenocytes isolated from APOE2/E2, APOE3/E3, or APOE4/E4 mice following L. monocytogenes infection. In vivo, overall susceptibility to foodborne listeriosis also did not differ by APOE genotype in either young (2 mo old) or aged (15 mo old) C57BL/6 mice. However, we observed a sex-dependent susceptibility to infection in aged APOE2/E2 male mice and a sex-dependent resistance to infection in aged APOE4/E4 male mice that was not present in female mice. Thus, these results suggest that APOE genotype does not play an important role in innate resistance to infection with L. monocytogenes but may be linked to sex-dependent changes that occur during immune senescence.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Feminino , Humanos , Masculino , Camundongos , Apolipoproteína E2 , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Citocinas , Genótipo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Alzheimers Dement ; 18(10): 1721-1735, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34908231

RESUMO

N-linked protein glycosylation in the brain is an understudied facet of glucose utilization that impacts a myriad of cellular processes including resting membrane potential, axon firing, and synaptic vesicle trafficking. Currently, a spatial map of N-linked glycans within the normal and Alzheimer's disease (AD) human brain does not exist. A comprehensive analysis of the spatial N-linked glycome would improve our understanding of brain energy metabolism, linking metabolism to signaling events perturbed during AD progression, and could illuminate new therapeutic strategies. Herein we report an optimized in situ workflow for enzyme-assisted, matrix-assisted laser desorption and ionization (MALDI) mass spectrometry imaging (MSI) of brain N-linked glycans. Using this workflow, we spatially interrogated N-linked glycan heterogeneity in both mouse and human AD brains and their respective age-matched controls. We identified robust regional-specific N-linked glycan changes associated with AD in mice and humans. These data suggest that N-linked glycan dysregulation could be an underpinning of AD pathologies.


Assuntos
Doença de Alzheimer , Glicômica , Humanos , Glicômica/métodos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Encéfalo/metabolismo , Polissacarídeos/análise , Polissacarídeos/química , Polissacarídeos/metabolismo , Glucose/metabolismo
4.
Alzheimers Dement ; 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36479795

RESUMO

Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.

5.
Nano Lett ; 20(8): 6135-6141, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32628854

RESUMO

We present the application of multiphoton in vivo fluorescence correlation spectroscopy (FCS) of fluorescent nanoparticles for the measurement of cerebral blood flow with excellent spatial and temporal resolution. Through the detection of single nanoparticles within the complex vessel architecture of a live mouse, this new approach enables the quantification of nanoparticle dynamics occurring within the vasculature along with simultaneous measurements of blood flow properties in the brain. In addition to providing high resolution blood flow measurements, this approach enables real-time quantification of nanoparticle concentration, degradation, and transport. This method is capable of quantifying flow rates at each pixel with submicron resolution to enable monitoring of dynamic changes in flow rates in response to changes in the animal's physiological condition. Scanning the excitation beam using FCS provides pixel by pixel mapping of flow rates with subvessel resolution across capillaries 300 µm deep in the brains of mice.


Assuntos
Microscopia de Fluorescência por Excitação Multifotônica , Nanopartículas , Animais , Circulação Cerebrovascular , Camundongos , Espectrometria de Fluorescência
6.
Neurobiol Dis ; 138: 104809, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32087284

RESUMO

Apolipoprotein E (APOE) is a multifunctional protein synthesized and secreted by multiple mammalian tissues. Although hepatocytes contribute about 75% of the peripheral pool, APOE can also be expressed in adipose tissue, the kidney, and the adrenal glands, among other tissues. High levels of APOE production also occur in the brain, where it is primarily synthesized by glia, and peripheral and brain APOE pools are thought to be distinct. In humans, APOE is polymorphic, with three major alleles (ε2, ε3, and ε4). These allelic forms dramatically alter APOE structure and function. Historically, the vast majority of research on APOE has centered on the important role it plays in modulating risk for cardiovascular disease and Alzheimer's disease. However, the established effects of this pleiotropic protein extend well beyond these two critical health challenges, with demonstrated roles across a wide spectrum of biological conditions, including adipose tissue function and obesity, metabolic syndrome and diabetes, fertility and longevity, and immune function. While the spectrum of biological systems in which APOE plays a role seems implausibly wide at first glance, there are some potential unifying mechanisms that could tie these seemingly disparate disorders together. In the current review, we aim to concisely summarize a wide breadth of APOE-associated pathologies and to analyze the influence of APOE in the development of several distinct disorders in order to provide insight into potential shared mechanisms implied in these various pathophysiological processes.


Assuntos
Apolipoproteínas E/genética , Alelos , Encéfalo/metabolismo , Diabetes Mellitus , Feminino , Genótipo , Humanos , Longevidade/genética , Masculino , Síndrome Metabólica/genética , Obesidade/genética
7.
Neurobiol Dis ; 136: 104742, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31931141

RESUMO

The Apolipoprotein E (APOE) gene is a major genetic risk factor associated with Alzheimer's disease (AD). APOE encodes for three main isoforms in humans (E2, E3, and E4). Homozygous E4 individuals have more than a 10-fold higher risk for developing late-onset AD, while E2 carriers are protected. A hallmark of AD is a reduction in cerebral glucose metabolism, alluding to a strong metabolic component in disease onset and progression. Interestingly, E4 individuals display a similar regional pattern of cerebral glucose hypometabolism decades prior to disease onset. Mapping this metabolic landscape may help elucidate the underlying biological mechanism of APOE-associated risk for AD. Efficient metabolic coupling of neurons and glia is necessary for proper neuronal function, and disruption in glial energy distribution has been proposed to contribute to neuronal cell death and AD pathology. One important function of astrocytes - canonically the primary source of apolipoprotein E in the brain - is to provide metabolic substrates (lactate, lipids, amino acids and neurotransmitters) to neurons. Here we investigate the effects of APOE on astrocyte glucose metabolism in vitro utilizing scintillation proximity assays, stable isotope tracer metabolomics, and gene expression analyses. Glucose uptake is impaired in E4 astrocytes relative to E2 or E3 with specific alterations in central carbon metabolism. Using stable isotope labeled glucose [U-13C] allowed analyses of astrocyte-specific deep metabolic networks affected by APOE, and provided insight to the effects downstream of glucose uptake. Enrichment of 13C in early steps of glycolysis was lowest in E4 astrocytes (highest in E2), while synthesis of lactate from glucose was highest in E4 astrocytes (lowest in E2). We observed an increase in glucose flux through the pentose phosphate pathway (PPP), with downstream increases in gluconeogenesis, lipid, and de novo nucleotide biosynthesis in E4 astrocytes. There was also a marked increase in 13C enrichment in the TCA cycle of E4 astrocytes - whose substrates were also incorporated into biosynthetic pathways at a higher rate. Pyruvate carboxylase (PC) and pyruvate dehydrogenase (PDH) are the two main enzymes controlling pyruvate entry to the TCA cycle. PC gene expression is increased in E4 astrocytes and the activity relative to PDH was also increased, compared to E2 or E3. Decreased enrichment in the TCA cycle of E2 and E3 astrocytes is suggestive of increased oxidation and non-glucose derived anaplerosis, which could be fueling mitochondrial ATP production. Conversely, E4 astrocytes appear to increase carbon flux into the TCA cycle to fuel cataplerosis. Together, these data demonstrate clear APOE isoform-specific effects on glucose utilization in astrocytes, including E4-associated increases in lactate synthesis, PPP flux, and de novo biosynthesis pathways.


Assuntos
Apolipoproteína E4/metabolismo , Astrócitos/metabolismo , Isótopos de Carbono/metabolismo , Glucose/metabolismo , Animais , Apolipoproteína E4/genética , Astrócitos/química , Isótopos de Carbono/análise , Linhagem Celular Transformada , Cromatografia por Troca Iônica/métodos , Glucose/análise , Humanos , Camundongos
8.
Curr Opin Lipidol ; 30(1): 10-15, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550413

RESUMO

PURPOSE OF REVIEW: Various groups have explored the effect of apolipoprotein E (APOE) on neurodegeneration through nutritional and metabolic alterations. In this review, we hope to summarize recent findings in humans as well as preclinical APOE models. RECENT FINDINGS: Metabolic pathways including lipid metabolism appear to play a large role in the pathophysiology of Alzheimer's disease. Carrier status of the E4 variant of the APOE gene is the strongest genetic risk factor for Alzheimer's disease, and increasing evidence suggests that E4 carriers may respond differently to a host of dietary and metabolic-related treatments. A new appreciation is forming for the role of APOE in cerebral metabolism, and how nutritional factors may impact this role. SUMMARY: Considering the role dietary factors play in APOE-associated cognitive decline will help us to understand how nutritional interventions may facilitate or mitigate disease progression.


Assuntos
Apolipoproteínas E/metabolismo , Demência/metabolismo , Nutrientes/metabolismo , Animais , Encéfalo/metabolismo , Dieta , Humanos
11.
FASEB J ; 28(8): 3769-79, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24812087

RESUMO

Impaired adipogenesis renders an adipose tissue unable to expand, leading to lipotoxicity and conditions such as diabetes and cardiovascular disease. While factors important for adipogenesis have been studied extensively, those that set the limits of adipose tissue expansion remain undetermined. Feeding a Western-type diet to apolipoprotein E2 knock-in mice, a model of metabolic syndrome, produced 3 groups of equally obese mice: mice with normal glucose tolerance, hyperinsulinemic yet glucose-tolerant mice, and prediabetic mice with impaired glucose tolerance and reduced circulating insulin. Using proteomics, we compared subcutaneous adipose tissues from mice in these groups and found that the expression of PTRF (polymerase I and transcript release factor) associated selectively with their glucose tolerance status. Lentiviral and pharmacologically overexpressed PTRF, whose function is critical for caveola formation, compromised adipocyte differentiation of cultured 3T3-L1cells. In human adipose tissue, PTRF mRNA levels positively correlated with markers of lipolysis and cellular senescence. Furthermore, a negative relationship between telomere length and PTRF mRNA levels was observed in human subcutaneous fat. PTRF is associated with limited adipose tissue expansion underpinning the key role of caveolae in adipocyte regulation. Furthermore, PTRF may be a suitable adipocyte marker for predicting pathological obesity and inform clinical management.


Assuntos
Adipócitos/patologia , Adipogenia/fisiologia , Cavéolas/fisiologia , Dieta/efeitos adversos , Intolerância à Glucose/etiologia , Hiperinsulinismo/etiologia , Obesidade/etiologia , Estado Pré-Diabético/etiologia , Proteínas de Ligação a RNA/fisiologia , Gordura Subcutânea/metabolismo , Células 3T3-L1 , Adiponectina/sangue , Animais , Aorta/patologia , Apolipoproteína E2/genética , Senescência Celular , Feminino , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Intolerância à Glucose/sangue , Intolerância à Glucose/patologia , Humanos , Hiperinsulinismo/sangue , Hiperinsulinismo/patologia , Resistência à Insulina , Lipólise , Fígado/química , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/classificação , Obesidade/patologia , Estado Pré-Diabético/sangue , Estado Pré-Diabético/patologia , Gravidez , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Gordura Subcutânea/patologia , Encurtamento do Telômero , Triglicerídeos/metabolismo
12.
J Neurochem ; 129(3): 495-508, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24400874

RESUMO

Dysregulation of hypothalamic-pituitary-adrenal (HPA) axis activation is associated with changes in addiction-related behaviors. In this study, we tested whether sex differences in the acute effects of methamphetamine (MA) exposure involve differential activation of the HPA axis. Male and female mice were injected with MA (1 mg/kg) or saline for comparison of plasma corticosterone and analysis of the immediate early gene c-Fos in brain. There was a prolonged elevation in corticosterone levels in female compared to male mice. C-Fos was elevated in both sexes following MA in HPA axis-associated regions, including the hypothalamic paraventricular nucleus (PVN), central amygdala, cingulate, and CA3 hippocampal region. MA increased the number of c-Fos and c-Fos/glucocorticoid receptor (GR) dual-labeled cells to a greater extent in males than females in the cingulate and CA3 regions. MA also increased the number of c-fos/vasopressin dual-labeled cells in the PVN as well as the number and percentage of c-Fos/GR dual-labeled cells in the PVN and central amygdala, although no sex differences in dual labeling were found in these regions. Thus, sex differences in MA-induced plasma corticosterone levels and activation of distinct brain regions and proteins involved in HPA axis regulation may contribute to sex differences in acute effects of MA on the brain. Methamphetamine induces a prolonged plasma corticosterone response in females compared to males. This may be mediated by increased neural activation, involving a greater activation of glucocorticoid receptor-positive cells, in males in the CA3 and cingulate brain regions, which are involved in negative feedback functions. These findings indicate a sex difference in the neural regulation of methamphetamine-induced plasma corticosterone release.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Metanfetamina/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Caracteres Sexuais , Animais , Corticosterona/sangue , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Radioimunoensaio
13.
Neurobiol Dis ; 64: 150-62, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24412220

RESUMO

Human apolipoprotein E (apoE) exists in three isoforms: apoE2, apoE3 and apoE4. APOE ε4 is a major genetic risk factor for cardiovascular disease (CVD) and Alzheimer's disease (AD). ApoE mediates cholesterol metabolism by binding various receptors. The low-density lipoprotein receptor (LDLR) has a high affinity for apoE, and is the only member of its receptor family to demonstrate an apoE isoform specific binding affinity (E4>E3>>E2). Evidence suggests that a functional interaction between apoE and LDLR influences the risk of CVD and AD. We hypothesize that the differential cognitive effects of the apoE isoforms are a direct result of their varying interactions with LDLR. To test this hypothesis, we have employed transgenic mice that express human apoE2, apoE3, or apoE4, and either human LDLR (hLDLR) or no LDLR (LDLR(-/-)). Our results show that plasma and brain apoE levels, cortical cholesterol, and spatial memory are all regulated by isoform-dependent interactions between apoE and LDLR. Conversely, both anxiety-like behavior and cued associative memory are strongly influenced by APOE genotype, but these processes appear to occur via an LDLR-independent mechanism. Both the lack of LDLR and the interaction between E4 and the LDLR were associated with significant impairments in the retention of long term spatial memory. Finally, levels of hippocampal apoE correlate with long term spatial memory retention in mice with human LDLR. In summary, we demonstrate that the apoE-LDLR interaction affects regional brain apoE levels, brain cholesterol, and cognitive function in an apoE isoform-dependent manner.


Assuntos
Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Memória de Longo Prazo/fisiologia , Receptores de LDL/metabolismo , Percepção Espacial/fisiologia , Animais , Ansiedade/metabolismo , Apolipoproteína E2/sangue , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E3/sangue , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/sangue , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/sangue , Apolipoproteínas E/genética , Colesterol/sangue , Colesterol/metabolismo , Condicionamento Psicológico/fisiologia , Humanos , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de LDL/genética
14.
bioRxiv ; 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38464274

RESUMO

Metabolism plays an important role in the maintenance of vigilance states (e.g. wake, NREM, and REM). Brain lactate fluctuations are a biomarker of sleep. Increased interstitial fluid (ISF) lactate levels are necessary for arousal and wake-associated behaviors, while decreased ISF lactate is required for sleep. ATP-sensitive potassium (K ATP ) channels couple glucose-lactate metabolism with neuronal excitability. Therefore, we explored how deletion of neuronal K ATP channel activity (Kir6.2-/- mice) affected the relationship between glycolytic flux, neuronal activity, and sleep/wake homeostasis. Kir6.2-/- mice shunt glucose towards glycolysis, reduce neurotransmitter synthesis, dampen cortical EEG activity, and decrease arousal. Kir6.2-/- mice spent more time awake at the onset of the light period due to altered ISF lactate dynamics. Together, we show that Kir6.2-K ATP channels act as metabolic sensors to gate arousal by maintaining the metabolic stability of each vigilance state and providing the metabolic flexibility to transition between states. Highlights: Glycolytic flux is necessary for neurotransmitter synthesis. In its absence, neuronal activity is compromised causing changes in arousal and vigilance states despite sufficient energy availability. With Kir6.2-K ATP channel deficiency, the ability to both maintain and shift between different vigilance states is compromised due to changes in glucose utilization. Kir6.2-K ATP channels are metabolic sensors under circadian control that gate arousal and sleep/wake transitions.

15.
J Lipid Res ; 54(2): 386-96, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23204275

RESUMO

Diabetes is a major risk factor for cardiovascular disease. To examine how diabetes interacts with a mildly compromised lipid metabolism, we introduced the diabetogenic Ins2(C96Y/+) (Akita) mutation into mice expressing human apoE4 (E4) combined with either an overexpressing human LDL receptor gene (hLDLR) or the wild-type mouse gene. The hLDLR allele caused 2-fold reductions in plasma HDL-cholesterol, plasma apoA1, and hepatic triglyceride secretion. Diabetes increased plasma total cholesterol 1.3-fold and increased apoB48 secretion 3-fold, while reducing triglyceride secretion 2-fold. Consequently, diabetic E4 mice with hLDLR secrete increased numbers of small, cholesterol-enriched, apoB48-containing VLDL, although they have near normal plasma cholesterol (<120 mg/dl). Small foam cell lesions were present in the aortic roots of all diabetic E4 mice with hLDLR that we analyzed at six months of age. None were present in nondiabetic mice or in diabetic mice without hLDLR. Aortic expression of genes affecting leukocyte recruitment and adhesion was enhanced by diabetes. ApoA1 levels, but not diabetes, were strongly correlated with the ability of plasma to efflux cholesterol from macrophages. We conclude that the diabetes-induced proinflammatory changes in the vasculature and the hLDLR-mediated cholesterol accumulation in macrophages synergistically trigger atherosclerosis in mice with human apoE4, although neither alone is sufficient.


Assuntos
Apolipoproteína E4/genética , Aterosclerose/metabolismo , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Angiopatias Diabéticas/metabolismo , Lipoproteínas/metabolismo , Alelos , Animais , Aterosclerose/genética , Aterosclerose/patologia , Transporte Biológico , Colesterol/metabolismo , Complicações do Diabetes/genética , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/patologia , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Lipoproteínas/sangue , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptores de LDL/genética
16.
J Neurochem ; 125(2): 303-13, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23383735

RESUMO

Oxidative stress (OS) and reactive oxygen species (ROS) play a modulatory role in synaptic plasticity and signaling pathways. Mitochondria (MT), a major source of ROS because of their involvement in energy metabolism, are important for brain function. MT-generated ROS are proposed to be responsible for a significant proportion of OS and are associated with developmental abnormalities and aspects of cellular aging. The role of ROS and MT function in cognition of healthy individuals is relatively understudied. In this study, we characterized behavioral and cognitive performance of 5- to 6-month-old mice over-expressing mitochondrial catalase (MCAT). MCAT mice showed enhancements in hippocampus-dependent spatial learning and memory in the water maze and contextual fear conditioning, and reduced measures of anxiety in the elevated zero maze. Catalase activity was elevated in MCAT mice in all brain regions examined. Measures of oxidative stress (glutathione, protein carbonyl content, lipid peroxidation, and 8-hydroxyguanine) did not significantly differ between the groups. The lack of differences in these markers of oxidative stress suggests that the differences observed in this study may be due to altered redox signaling. Catalase over-expression might be sufficient to enhance cognition and reduce measures of anxiety even in the absence of alteration in levels of OS.


Assuntos
Ansiedade/enzimologia , Catalase/biossíntese , Hipocampo/metabolismo , Memória/fisiologia , Mitocôndrias/enzimologia , Animais , Ansiedade/fisiopatologia , Humanos , Imuno-Histoquímica , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Estresse Oxidativo/fisiologia
17.
Arterioscler Thromb Vasc Biol ; 32(6): 1436-44, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22539598

RESUMO

OBJECTIVE: The dominant-negative mutation, P467L, in peroxisome proliferator-activated receptor-γ (PPARγ) affects adipose tissue distribution, insulin sensitivity, and blood pressure in heterozygous humans. We hypothesized that the equivalent mutation, PPARγ-P465L, in mice will worsen atherosclerosis. METHODS AND RESULTS: Apolipoprotein E-null mice with and without PPARγ-P465L mutation were bred in 129S6 inbred genetic background. Mild hypertension and lipodystrophy of PPARγ-P465L persisted in the apolipoprotein E-null background. Glucose homeostasis was normal, but plasma adiponectin was significantly lower and resistin was higher in PPARγ-P465L mice. Plasma cholesterol and lipoprotein distribution were not different, but plasma triglycerides tended to be reduced. Surprisingly, there were no overall changes in the atherosclerotic plaque size or composition. PPARγ-P465L macrophages had a small decrease in CD36 mRNA and a small yet significant reduction in very-low-density lipoprotein uptake in culture. In unloaded apolipoprotein E-null macrophages with PPARγ-P465L, cholesterol uptake was reduced whereas apolipoprotein AI-mediated efflux was increased. However, when cells were cholesterol loaded in the presence of acetylated low-density lipoprotein, no genotype difference in uptake or efflux was apparent. A reduction of vascular cell adhesion molecule-1 expression in aorta suggests a relatively antiatherogenic vascular environment in mice with PPARγ-P465L. CONCLUSIONS: Small, competing pro- and antiatherogenic effects of PPARγ-P465L mutation result in unchanged plaque development in apolipoprotein E-deficient mice.


Assuntos
Doenças da Aorta/genética , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/genética , Aterosclerose/prevenção & controle , Mutação , PPAR gama/genética , Adiponectina/sangue , Animais , Doenças da Aorta/sangue , Doenças da Aorta/etiologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Glicemia/metabolismo , Transplante de Medula Óssea , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Colesterol/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genótipo , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , PPAR gama/metabolismo , Fenótipo , Resistina/sangue , Fatores de Tempo , Triglicerídeos/sangue , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
18.
bioRxiv ; 2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37609198

RESUMO

Background: Inflammatory cells within atherosclerotic lesions secrete various proteolytic enzymes that contribute to lesion progression and destabilization, increasing the risk for an acute cardiovascular event. The relative contributions of specific proteases to atherogenesis is not well understood. Elastase is a serine protease, secreted by macrophages and neutrophils, that may contribute to the development of unstable plaque. We have previously reported interaction of endogenous protease-inhibitor proteins with high-density lipoprotein (HDL), including alpha-1-antitrypsin, an inhibitor of elastase. These findings support a potential role for HDL as an endogenous modulator of protease activity. In this study, we test the hypothesis that enhancement of HDL-associated elastase inhibitor activity is protective against atherosclerotic lesion progression. Methods: We designed an HDL-targeting protease inhibitor (HTPI) that binds to HDL and confers elastase inhibitor activity. Lipoprotein binding and the impact of HTPI on atherosclerosis was examined using mouse models. Results: HTPI is a small (1.6 kDa) peptide with an elastase inhibitor domain, a soluble linker, and an HDL-targeting domain. When incubated with human plasma ex vivo , HTPI predominantly binds to HDL. Intravenous administration of HTPI to mice resulted in its binding to plasma HDL and increased elastase inhibitor activity on isolated HDL. Accumulation of HTPI within plaque was observed after systemic administration to Apoe -/- mice. To examine the effect of HTPI treatment on atherosclerosis, prevention and progression studies were performed using Ldlr -/- mice fed Western diet. In both study designs, HTPI-treated mice had reduced lipid deposition in plaque. Histology and immunofluorescence staining of aortic root sections were used to examine the impact of HTPI on lesion morphology and inflammatory features. Conclusions: These data support the hypothesis that HDL-associated anti-elastase activity can improve the atheroprotective potential of HDL and highlight the potential utility of HDL enrichment with anti-protease activity as an approach for stabilization of atherosclerotic lesions.

19.
ACS Nano ; 17(13): 12862-12874, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37341451

RESUMO

Unraveling the transport of drugs and nanocarriers in cerebrovascular networks is important for pharmacokinetic and hemodynamic studies but is challenging due to the complexity of sensing individual particles within the circulatory system of a live animal. Here, we demonstrate that a DNA-stabilized silver nanocluster (DNA-Ag16NC) that emits in the first near-infrared window upon two-photon excitation in the second NIR window can be used for multiphoton in vivo fluorescence correlation spectroscopy for the measurement of cerebral blood flow rates in live mice with high spatial and temporal resolution. To ensure bright and stable emission during in vivo experiments, we loaded DNA-Ag16NCs into liposomes, which served the dual purposes of concentrating the fluorescent label and protecting it from degradation. DNA-Ag16NC-loaded liposomes enabled the quantification of cerebral blood flow velocities within individual vessels of a living mouse.


Assuntos
DNA , Lipossomos , Animais , Camundongos , DNA/química , Corantes , Espectrometria de Fluorescência , Circulação Cerebrovascular , Corantes Fluorescentes/química
20.
Sci Rep ; 13(1): 16855, 2023 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803069

RESUMO

Mounting evidence highlights the crucial role of aging in the pathogenesis of Alzheimer's disease (AD). We have previously explored human apoE-targeted replacement mice across different ages and identified distinct molecular pathways driven by aging. However, the specific contribution of different brain cell types to the gene modules underlying these pathways remained elusive. To bridge this knowledge gap, we employed a computational deconvolution approach to examine cell-type-specific gene expression profiles in major brain cell types, including astrocytes (AS), microglia (MG), oligodendroglia (OG), neurons (NEU), and vascular cells (VC). Our findings revealed that immune module genes were predominantly expressed in MG, OG, and VC. The lipid metabolism module genes were primarily expressed in AS, MG, and OG. The mitochondria module genes showed prominent expression in VC, and the synapse module genes were primarily expressed in NEU and VC. Furthermore, we identified intra- and inter-cell-type interactions among these module genes and validated their aging-associated expression changes using published single cell studies. Our study dissected bulk brain transcriptomics data at the cellular level, providing a closer examination of the cell-type contributions to the molecular pathways driven by aging.


Assuntos
Doença de Alzheimer , Transcriptoma , Camundongos , Humanos , Animais , Perfilação da Expressão Gênica , Doença de Alzheimer/metabolismo , Envelhecimento/genética , Encéfalo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA