Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuron ; 109(10): 1657-1674.e7, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33831349

RESUMO

The apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease and directly influences tauopathy and tau-mediated neurodegeneration. ApoE4 has strong deleterious effects on both parameters. In the brain, apoE is produced and secreted primarily by astrocytes and by activated microglia. The cell-specific role of each form of apoE in the setting of neurodegeneration has not been determined. We generated P301S Tau/Aldh1l1-CreERT2/apoE3flox/flox or Tau/Aldh1l1-CreERT2/apoE4flox/flox mice. At 5.5 months of age, after the onset of tau pathology, we administered tamoxifen or vehicle and compared mice at 9.5 months of age. Removing astrocytic APOE4 markedly reduced tau-mediated neurodegeneration and decreased phosphorylated tau (pTau) pathology. Single-nucleus RNA sequencing analysis revealed striking gene expression changes in all cell types, with astrocytic APOE4 removal decreasing disease-associated gene signatures in neurons, oligodendrocytes, astrocytes, and microglia. Removal of astrocytic APOE4 decreased tau-induced synaptic loss and microglial phagocytosis of synaptic elements, suggesting a key role for astrocytic apoE in synaptic degeneration.


Assuntos
Apolipoproteína E4/metabolismo , Astrócitos/metabolismo , Fagocitose , Tauopatias/metabolismo , Animais , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Apoptose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Sinapses/metabolismo , Sinapses/patologia , Tauopatias/patologia , Transcriptoma , Proteínas tau/metabolismo
2.
Int J Mol Sci ; 21(4)2020 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-32075060

RESUMO

Apolipoprotein E (APOE) ε4 gene allele and type 2 diabetes mellitus (T2DM) are prime risk factors for Alzheimer's disease (AD). Despite evidence linking T2DM and apoE4, the mechanism underlying their interaction is yet to be determined. In the present study, we employed a model of APOE-targeted replacement mice and high-fat diet (HFD)-induced insulin resistance to investigate diabetic mechanisms associated with apoE4 pathology and the extent to which they are driven by peripheral and central processes. Results obtained revealed an intriguing pattern, in which under basal conditions, apoE4 mice display impaired glucose and insulin tolerance and decreased insulin secretion, as well as cognitive and sensorimotor characteristics relative to apoE3 mice, while the HFD impairs apoE3 mice without significantly affecting apoE4 mice. Measurements of weight and fasting blood glucose levels increased in a time-dependent manner following the HFD, though no effect of genotype was observed. Interestingly, sciatic electrophysiological and skin intra-epidermal nerve fiber density (IENFD) peripheral measurements were not affected by the APOE genotype or HFD, suggesting that the observed sensorimotor and cognitive phenotypes are related to central nervous system processes. Indeed, measurements of hippocampal insulin receptor and glycogen synthase kinase-3ß (GSK-3ß) activation revealed a pattern similar to that obtained in the behavioral measurements while Akt activation presented a dominant effect of diet. HFD manipulation induced genotype-independent hyperlipidation of apoE, and reduced levels of brain apoE in apoE3 mice, rendering them similar to apoE4 mice, whose brain apoE levels were not affected by the diet. No such effect was observed in the peripheral plasma levels of apoE, suggesting that the pathological effects of apoE4 under the control diet and apoE3 under HFD conditions are related to the decreased levels of brain apoE. Taken together, our data suggests that diabetic mechanisms play an important role in mediating the pathological effects of apoE4 and that consequently, diabetic-related therapy may be useful in treating apoE4 pathology in AD.


Assuntos
Apolipoproteína E4/metabolismo , Diabetes Mellitus Tipo 2/patologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Apolipoproteínas E/sangue , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Dieta Hiperlipídica , Genótipo , Teste de Tolerância a Glucose , Hipocampo/metabolismo , Humanos , Locomoção , Memória de Curto Prazo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Medição da Dor , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
3.
Nature ; 549(7673): 523-527, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28959956

RESUMO

APOE4 is the strongest genetic risk factor for late-onset Alzheimer disease. ApoE4 increases brain amyloid-ß pathology relative to other ApoE isoforms. However, whether APOE independently influences tau pathology, the other major proteinopathy of Alzheimer disease and other tauopathies, or tau-mediated neurodegeneration, is not clear. By generating P301S tau transgenic mice on either a human ApoE knock-in (KI) or ApoE knockout (KO) background, here we show that P301S/E4 mice have significantly higher tau levels in the brain and a greater extent of somatodendritic tau redistribution by three months of age compared with P301S/E2, P301S/E3, and P301S/EKO mice. By nine months of age, P301S mice with different ApoE genotypes display distinct phosphorylated tau protein (p-tau) staining patterns. P301S/E4 mice develop markedly more brain atrophy and neuroinflammation than P301S/E2 and P301S/E3 mice, whereas P301S/EKO mice are largely protected from these changes. In vitro, E4-expressing microglia exhibit higher innate immune reactivity after lipopolysaccharide treatment. Co-culturing P301S tau-expressing neurons with E4-expressing mixed glia results in a significantly higher level of tumour-necrosis factor-α (TNF-α) secretion and markedly reduced neuronal viability compared with neuron/E2 and neuron/E3 co-cultures. Neurons co-cultured with EKO glia showed the greatest viability with the lowest level of secreted TNF-α. Treatment of P301S neurons with recombinant ApoE (E2, E3, E4) also leads to some neuronal damage and death compared with the absence of ApoE, with ApoE4 exacerbating the effect. In individuals with a sporadic primary tauopathy, the presence of an ε4 allele is associated with more severe regional neurodegeneration. In individuals who are positive for amyloid-ß pathology with symptomatic Alzheimer disease who usually have tau pathology, ε4-carriers demonstrate greater rates of disease progression. Our results demonstrate that ApoE affects tau pathogenesis, neuroinflammation, and tau-mediated neurodegeneration independently of amyloid-ß pathology. ApoE4 exerts a 'toxic' gain of function whereas the absence of ApoE is protective.


Assuntos
Apolipoproteína E4/metabolismo , Apolipoproteína E4/toxicidade , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/metabolismo , Alelos , Animais , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Introdução de Genes , Genótipo , Humanos , Imunidade Inata , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microglia/imunologia , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fosfoproteínas/análise , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Tauopatias/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas tau/genética
4.
PLoS One ; 11(5): e0155307, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171180

RESUMO

Apolipoprotein E4 (ApoE4), one of three common isoforms of ApoE, is a major risk factor for late-onset Alzheimer disease (AD). ApoE-deficient mice, as well as mice expressing human ApoE4, display impaired learning and memory functions and signs of neurodegeneration. Moreover, ApoE protects against high-fat (HF) diet induced neurodegeneration by its role in the maintenance of the integrity of the blood-brain barrier. The influence of a HF diet on the progression of AD-like cognitive and neuropathological changes was assessed in wild-type (WT), human ApoE4 and ApoE-knockout (ApoE-/-) mice to evaluate the modulatory role of ApoE in this process. From 12 months of age, female WT, ApoE4, and ApoE-/- mice were fed either a standard or a HF diet (19% butter, 0.5% cholate, 1.25% cholesterol) throughout life. At 15 months of age mice performed the Morris water maze, evaluating spatial learning and memory. ApoE-/- showed increased spatial learning compared to WT mice (p = 0.009). HF diet improved spatial learning in WT mice (p = 0.045), but did not affect ApoE4 and ApoE-/- mice. Immunohistochemical analyses of the hippocampus demonstrated increased neuroinflammation (CD68) in the cornu ammonis 1 (CA1) region in ApoE4 (p = 0.001) and in ApoE-/- (p = 0.032) mice on standard diet. HF diet tended to increase CD68 in the CA1 in WT mice (p = 0.052), while it decreased in ApoE4 (p = 0.009), but ApoE-/- remained unaffected. A trend towards increased neurogenesis (DCX) was found in both ApoE4 (p = 0.052) and ApoE-/- mice (p = 0.068). In conclusion, these data suggest that HF intake induces different effects in WT mice compared to ApoE4 and ApoE-/- with respect to markers for cognition and neurodegeneration. We propose that HF intake inhibits the compensatory mechanisms of neuroinflammation and neurogenesis in aged female ApoE4 and ApoE-/- mice.


Assuntos
Apolipoproteína E4/deficiência , Encéfalo/patologia , Encéfalo/fisiopatologia , Cognição/fisiologia , Inflamação/patologia , Plasticidade Neuronal , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apolipoproteína E4/metabolismo , Peso Corporal , Giro Denteado/metabolismo , Dieta Hiperlipídica , Proteína 4 Homóloga a Disks-Large , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Técnicas de Introdução de Genes , Transportador de Glucose Tipo 1/metabolismo , Guanilato Quinases/metabolismo , Humanos , Imuno-Histoquímica , Aprendizagem em Labirinto , Proteínas de Membrana/metabolismo , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Tamanho do Órgão
5.
Nat Commun ; 7: 10918, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26948379

RESUMO

Increased APP (amyloid precursor protein) processing causes ß-amyloid (Aß) accumulation in autosomal dominant Alzheimer's disease (AD), but it is unclear if it also affects sporadic Aß accumulation. We tested healthy controls and patients with mild cognitive symptoms (N=331) in the BioFINDER study, using cerebrospinal fluid (CSF) Aß40 as a surrogate for amyloidogenic APP processing. We find that levels of brain Aß fibrils (measured by 18F-flutemetamol PET) are independently associated with high CSF Aß40 (P<0.001) and APOE ɛ4 (P<0.001). The association between CSF Aß40 and brain Aß is stronger in APOE ɛ4-negative than in positive people (P=0.0080). The results are similar for CSF Aß38 and for a combination of CSF Aß38 and CSF Aß40. In conclusion, sporadic Aß accumulation may be partly associated with increased amyloidogenic APP production, especially in APOE ɛ4-negative subjects. The risk for sporadic AD may consequently depend on increased Aß production, in addition to decreased Aß clearance.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Apolipoproteína E4/deficiência , Idoso , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Amiloidose/genética , Apolipoproteína E4/genética , Encéfalo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
6.
J Viral Hepat ; 23(1): 39-46, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26306786

RESUMO

Hepatitis C virus (HCV) causes not only liver damage in certain patients but can also lead to neuropsychiatric symptoms. Previous studies have shown that the type 4 allele of the gene for apolipoprotein E (APOE) is strongly protective against HCV-induced damage in liver. In this study, we have investigated the possibility that APOE genotype is involved in the action of HCV in brain. One hundred HCV-infected patients with mild liver disease underwent a neurological examination and a comprehensive psychometric testing of attention and memory function. In addition, patients completed questionnaires for the assessment of fatigue, health-related quality of life and mood disturbances. Apolipoprotein E gene genotyping was carried out on saliva using buccal swabs. The APOE-ε4 allele frequency was significantly lower in patients with an impairment of working memory, compared to those with a normal working memory test result (P = 0.003). A lower APOE-ε4 allele frequency was also observed in patients with definitely altered attention ability (P = 0.008), but here, the P-value missed the level of significance after application of the Bonferroni correction. Our data suggest that the APOE-ε4 allele is protective against attention deficit and especially against poor working memory in HCV-infected subjects with mild liver disease. Considering the role of apolipoprotein E in the life cycle of the virus, the findings shed interesting new light upon possible pathomechanisms behind the development of neuropsychiatric symptoms in hepatitis C infection.


Assuntos
Apolipoproteína E4/deficiência , Disfunção Cognitiva/psicologia , Encefalopatia Hepática/psicologia , Hepatite C Crônica/patologia , Memória de Curto Prazo/fisiologia , Transtornos do Humor/psicologia , Doenças Neurodegenerativas/psicologia , Adulto , Idoso , Alelos , Apolipoproteína E4/genética , Cognição , Disfunção Cognitiva/virologia , Feminino , Frequência do Gene/genética , Hepacivirus/genética , Encefalopatia Hepática/virologia , Hepatite C Crônica/virologia , Humanos , Fígado/patologia , Fígado/virologia , Masculino , Pessoa de Meia-Idade , Transtornos do Humor/virologia , Doenças Neurodegenerativas/virologia , Testes Neuropsicológicos , Qualidade de Vida/psicologia , Inquéritos e Questionários
7.
J Neurosci ; 34(42): 13963-75, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25319693

RESUMO

It is well established that the cholesterol-transporter apolipoprotein ε (APOE) genotype is associated with the risk of developing neurodegenerative diseases. Recently, brain functional connectivity (FC) in apoE-ε4 carriers has been investigated by means of resting-state fMRI, showing a marked differentiation in several functional networks at different ages compared with carriers of other apoE isoforms. The causes of such hampered FC are not understood. We hypothesize that vascular function and synaptic repair processes, which are both impaired in carriers of ε4, are the major contributors to the loss of FC during aging. To test this hypothesis, we integrated several different MRI techniques with immunohistochemistry and investigated FC changes in relation with perfusion, diffusion, and synaptic density in apoE4 and apoE-knock-out (KO) mice at 12 (adult) and 18 months of age. Compared with wild-type mice, we detected FC deficits in both adult and old apoE4 and apoE-KO mice. In apoE4 mice, these changes occurred concomitant with increased mean diffusivity in the hippocampus, whereas perfusion deficits appear only later in life, together with reduced postsynaptic density levels. Instead, in apoE-KO mice FC deficits were mirrored by strongly reduced brain perfusion since adulthood. In conclusion, we provide new evidence for a relation between apoE and brain connectivity, possibly mediated by vascular risk factors and by the efficiency of APOE as synaptic modulator in the brain. Our results show that multimodal MR neuroimaging is an excellent tool to assess brain function and to investigate early neuropathology and aging effects in translational research.


Assuntos
Envelhecimento/metabolismo , Apolipoproteína E4/deficiência , Encéfalo/metabolismo , Rede Nervosa/metabolismo , Descanso/fisiologia , Envelhecimento/patologia , Animais , Apolipoproteínas E/deficiência , Encéfalo/patologia , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Rede Nervosa/patologia
8.
Curr Alzheimer Res ; 11(3): 283-90, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24694076

RESUMO

Apolipoprotein E ε4 allele (ApoE4) has been associated with increased risk of sporadic Alzheimer's disease (AD) and of conversion from mild cognitive impairment to AD. But the underlying mechanism of ApoE4 affecting brain atrophy and cognition is not fully understood. We investigated the effect of ApoE4 on amyloid beta (Aß) protein burden and its correlation with the structure change of hippocampus and cortex, cognitive and behavioral changes in ApoE4 transgenic mice. Male ApoE4 transgenic mice and age-matched control mice at age 12 months and 24 months were tested in the Morris Water Maze (MWM). Brain volume changes (including whole brain, hippocampus, cortex, total ventricles and caudate putamen) were assessed by using small animal 7T-MRI. Aß level was assessed by immunohistochemistry (IHC) and immunoprecipitation/western blot. In MWM, escape latency was longer and time spent in the target quadrant was shorter in aged ApoE4 mice (12- and 24-month-old), suggesting age- and ApoE4-dependent visuospatial deficits. Atrophy on MRI was prominent in the hippocampus (p=0.039) and cortex (p=0.013) of ApoE4 mice (24-month-old) as compared to age-matched control mice. IHC revealed elevated Aß deposition in the hippocampus. Consistently, both soluble and insoluble Aß aggregates were increased in aged ApoE4 mice. This increase was correlated inversely with hippocampal atrophy and cognitive deficits. These data give further evidence that ApoE4 plays an important role in brain atrophy and memory impairment by modulating amyloid production and deposition.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E4/deficiência , Encéfalo/patologia , Transtornos da Memória , Fragmentos de Peptídeos/metabolismo , Envelhecimento/genética , Animais , Apolipoproteína E4/genética , Atrofia/etiologia , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polissonografia , Tempo de Reação/genética , Estatísticas não Paramétricas , Fatores de Tempo
9.
Sci Rep ; 4: 3754, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24435134

RESUMO

Human ApoE4 accelerates memory decline in ageing and in Alzheimer's disease. Although intranasal insulin can improve cognition, this has little effect in ApoE4 subjects. To understand this ApoE genotype-dependent effect, we examined brain insulin signaling in huApoE3 and huApoE4 targeted replacement (TR) mice. At 32 weeks, lower insulin receptor substrate 1 (IRS1) at S636/639 and Akt phosphorylation at T308 were detected in fasting huApoE4 TR mice as compared to fasting huApoE3 TR mice. These changes in fasting huApoE4 TR mice were linked to lower brain glucose content and have no effect on plasma glucose level. However, at 72 weeks of age, these early changes were accompanied by reduction in IRS2 expression, IRS1 phosphorylation at Y608, Akt phosphorylation at S473, and MAPK (p38 and p44/42) activation in the fasting huApoE4 TR mice. The lower brain glucose was significantly associated with higher brain insulin in the aged huApoE4 TR mice. These results show that ApoE4 reduces brain insulin signaling and glucose level leading to higher insulin content.


Assuntos
Apolipoproteína E4/genética , Encéfalo/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apolipoproteína E4/deficiência , Glicemia , Colesterol/sangue , Colesterol/metabolismo , Expressão Gênica , Marcação de Genes , Glucose/metabolismo , Humanos , Insulina/sangue , Insulina/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
10.
Brain Struct Funct ; 219(5): 1841-68, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23832599

RESUMO

Lipid metabolism and genetic background together strongly influence the development of both cardiovascular and neurodegenerative diseases like Alzheimer's disease (AD). A non-pharmacological way to prevent the genotype-induced occurrence of these pathologies is given by dietary behavior. In the present study, we tested the effects of long-term consumption of a specific multi-nutrient diet in two models for atherosclerosis and vascular risk factors in AD: the apolipoprotein ε4 (apoE4) and the apoE knockout (apoE ko) mice. This specific multi-nutrient diet was developed to support neuronal membrane synthesis and was expected to contribute to the maintenance of vascular health. At 12 months of age, both genotypes showed behavioral changes compared to control mice and we found increased neurogenesis in apoE ko mice. The specific multi-nutrient diet decreased anxiety-related behavior in the open field, influenced sterol composition in serum and brain tissue, and increased the concentration of omega-3 fatty acids in the brain. Furthermore, we found that wild-type and apoE ko mice fed with this multi-nutrient diet showed locally increased cerebral blood volume and decreased hippocampal glutamate levels. Taken together, these data suggest that a specific dietary intervention has beneficial effects on early pathological consequences of hypercholesterolemia and vascular risk factors for AD.


Assuntos
Doença de Alzheimer/dietoterapia , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Encéfalo/metabolismo , Transtornos Cognitivos/prevenção & controle , Dieta , Hemodinâmica/genética , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Comportamento Exploratório , Hemodinâmica/efeitos dos fármacos , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurogênese , Tamanho do Órgão , Presenilina-1/genética
11.
Arterioscler Thromb Vasc Biol ; 33(4): 687-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23413428

RESUMO

OBJECTIVE: The goal of this study was to understand the molecular basis of how the amino acid substitution C112R that distinguishes human apolipoprotein (apo) E4 from apoE3 causes the more proatherogenic plasma lipoprotein-cholesterol distribution that is known to be associated with the expression of apoE4. APPROACH AND RESULTS: Adeno-associated viruses, serotype 8 (AAV8), were used to express different levels of human apoE3, apoE4, and several C-terminal truncation and internal deletion variants in C57BL/6 apoE-null mice, which exhibit marked dysbetalipoproteinemia. Plasma obtained from these mice 2 weeks after the AAV8 treatment was analyzed for cholesterol and triglyceride levels, as well as for the distribution of cholesterol between the lipoprotein fractions. Hepatic expression of apoE3 and apoE4 induced similar dose-dependent decreases in plasma cholesterol and triglyceride to the levels seen in control C57BL/6 mice. Importantly, at the same reduction in plasma total cholesterol, expression of apoE4 gave rise to higher very low-density lipoprotein-cholesterol (VLDL-C) and lower high-density lipoprotein-cholesterol levels relative to the apoE3 situation. The C-terminal domain and residues 261 to 272 in particular play a critical role, because deleting them markedly affected the performance of both isoforms. CONCLUSIONS: ApoE4 possesses enhanced lipid and VLDL-binding ability relative to apoE3, which gives rise to impaired lipolytic processing of VLDL in apoE4-expressing mice. These effects reduce VLDL remnant clearance from the plasma compartment and decrease the amount of VLDL surface components available for incorporation into the high-density lipoprotein pool, accounting for the more proatherogenic lipoprotein profile (higher VLDL-C/high-density lipoprotein-cholesterol ratio) occurring in apoE4-expressing animals compared with their apoE3 counterparts.


Assuntos
Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , HDL-Colesterol/sangue , VLDL-Colesterol/sangue , Colesterol/sangue , Hiperlipoproteinemia Tipo III/sangue , Substituição de Aminoácidos , Animais , Apolipoproteína E3/química , Apolipoproteína E3/deficiência , Apolipoproteína E3/genética , Apolipoproteína E4/química , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Hiperlipoproteinemia Tipo III/genética , Lipólise , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estrutura Terciária de Proteína , Fatores de Tempo , Triglicerídeos/sangue
12.
Nature ; 485(7399): 512-6, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22622580

RESUMO

Human apolipoprotein E has three isoforms: APOE2, APOE3 and APOE4. APOE4 is a major genetic risk factor for Alzheimer's disease and is associated with Down's syndrome dementia and poor neurological outcome after traumatic brain injury and haemorrhage. Neurovascular dysfunction is present in normal APOE4 carriers and individuals with APOE4-associated disorders. In mice, lack of Apoe leads to blood-brain barrier (BBB) breakdown, whereas APOE4 increases BBB susceptibility to injury. How APOE genotype affects brain microcirculation remains elusive. Using different APOE transgenic mice, including mice with ablation and/or inhibition of cyclophilin A (CypA), here we show that expression of APOE4 and lack of murine Apoe, but not APOE2 and APOE3, leads to BBB breakdown by activating a proinflammatory CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes. This, in turn, leads to neuronal uptake of multiple blood-derived neurotoxic proteins, and microvascular and cerebral blood flow reductions. We show that the vascular defects in Apoe-deficient and APOE4-expressing mice precede neuronal dysfunction and can initiate neurodegenerative changes. Astrocyte-secreted APOE3, but not APOE4, suppressed the CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes through a lipoprotein receptor. Our data suggest that CypA is a key target for treating APOE4-mediated neurovascular injury and the resulting neuronal dysfunction and degeneration.


Assuntos
Apolipoproteínas E/metabolismo , Barreira Hematoencefálica/fisiologia , Circulação Cerebrovascular/fisiologia , Ciclofilina A/metabolismo , Animais , Apolipoproteína E2/deficiência , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E3/deficiência , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/deficiência , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Microcirculação , NF-kappa B/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Pericitos/metabolismo
13.
J Neurosci ; 32(14): 4803-11, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492035

RESUMO

Apolipoprotein E4 (apoE4) plays a major role in the pathogenesis of Alzheimer's disease. Brain amyloid-ß (Aß) accumulation depends on age and apoE isoforms (apoE4 > apoE3) both in humans and in transgenic mouse models. Brain apoE levels are also isoform dependent, but in the opposite direction (apoE4 < apoE3). Thus, one prevailing hypothesis is to increase brain apoE expression to reduce Aß levels. To test this hypothesis, we generated mutant human amyloid precursor protein transgenic mice expressing one or two copies of the human APOE3 or APOE4 gene that was knocked in and flanked by LoxP sites. We report that reducing apoE3 or apoE4 expression by 50% in 6-month-old mice results in efficient Aß clearance and does not increase Aß accumulation. However, 12-month-old mice with one copy of the human APOE gene had significantly reduced Aß levels and plaque loads compared with mice with two copies, regardless of which human apoE isoform was expressed, suggesting a gene dose-dependent effect of apoE on Aß accumulation in aged mice. Additionally, 12-month-old mice expressing one or two copies of the human APOE4 gene had significantly higher levels of Aß accumulation and plaque loads than age-matched mice expressing one or two copies of the human APOE3 gene, suggesting an isoform-dependent effect of apoE on Aß accumulation in aged mice. Moreover, Cre-mediated APOE4 gene excision in hippocampal astrocytes significantly reduced insoluble Aß in adult mice. Thus, reducing, rather than increasing, apoE expression is an attractive approach to lowering brain Aß levels.


Assuntos
Envelhecimento/genética , Precursor de Proteína beta-Amiloide/genética , Apolipoproteína E3/deficiência , Apolipoproteína E4/deficiência , Apolipoproteínas E/deficiência , Mutação/genética , Envelhecimento/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/fisiologia , Feminino , Técnicas de Introdução de Genes , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
15.
J Alzheimers Dis ; 27(1): 89-98, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21743131

RESUMO

Apolipoprotein E ε4 (ApoE4) has been implicated as a potential genetic risk factor for dementia. In this study, we investigate the effect of ApoE4 on learning and memory, changes in brain volume and neuroinflammatory responses in brain of ApoE4 transgenic mice. Four groups of male mice with ApoE4 and age-matched wild type (WT) (6-, 12-, 18- and 24-month) were studied. Spatial learning and retaining of mice was examined in the Morris Water Maze (MWM). Changes in brain volume (including the whole brain, hippocampus, cortex, total ventricles, and caudate putamen) were assessed by using 7T small animal MRI. Neuroinflammatory responses were analyzed by measuring the levels of microglia (Iba-1), iNOS, TNFα, and IL-6 quantitatively. In the MWM, ApoE4 mice showed longer escape latency (p < 0.05) and swim distance (p < 0.05) at age 12 month and older, comparing with the WT mice. They also demonstrated poor memory retention in the probe test (p < 0.05). Brain atrophy was significant in ApoE4 mice than age-matched WT mice (18 months: 0.079 ± 0.004 versus 0.086 ± 0.003, p = 0.018; and 24 months: 0.074 ± 0.005 versus 0.084 ± 0.006, p = 0.008). The expression of Iba-1, iNOS, and TNFα in hippocampus and cortex were significantly higher in ApoE4 mice than in WT mice at 12 months and older. These data suggest that ApoE4 plays an important role in learning and memory impairment. These deficits are associated with neuroinflammatory responses that may in turn lead to atrophy in hippocampus and cortex.


Assuntos
Envelhecimento/patologia , Apolipoproteína E4/genética , Hipocampo/patologia , Deficiências da Aprendizagem/patologia , Memória/fisiologia , Fatores Etários , Análise de Variância , Animais , Apolipoproteína E4/deficiência , Humanos , Deficiências da Aprendizagem/genética , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Percepção Espacial
16.
Neurosci Bull ; 25(4): 167-78, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19633698

RESUMO

OBJECTIVE: Apolipoprotein E (apoE) is associated with increased risk of age-related diseases, such as Alzheimer's disease (AD) and cerebrovascular disease (CVD). The present study aims to investigate the age-related general morphological changes of the brain in GFAP-apoE transgenic mice, especially the alterations in number and size of hippocampal pyramidal cells and the microvascular lesions in the thalamus. METHODS: Nine female apoE4/4 mice were divided into 3 groups (n=3 in each group): 3-4 months (young group), 9-10 months (middle-aged group) and 20-21 months (old group). Age-matched apoE3/3 mice were employed as control group (n=3 in each group). The paraffin sections of brain tissue were stained by 2 conventional staining methods, thionin staining and hematoxylin-esion(HE) staining, the former of which was to observe the hippocampal cells, while the latter was used to examine the brain microvasculature. RESULTS: There was no apparent difference in the cortical layer between apoE3/3 and apoE4/4 mice, neither any significant difference in the number of cells in hippocampal CA1-CA3 subfields between apoE3/3 and apoE4/4 mice at various age points (P>0.05). However, the mean size of pyramidal cells in CA1 subfield in apoE3/3 and apoE4/4 mice decreased as mice were getting older (P<0.001). At the age of 20-21 months, this cellular atrophy in apoE4/4 mice was more severe than that in old apoE3/3 mice (P<0.05). Furthermore, microvascular lesion in the thalamus was detected in all the 3 old apoE4/4 mice, at varying degrees (5.24%, 1.41% and 3.97%, respectively), while only one apoE3/3 mouse exhibited microvascular lesion in the thalamus, at a low level (0.85%). CONCLUSION: The current study suggests that the cell size in hippocampal CA1 subfield decreases with aging, irrespective of apoE genotype. Cellular atrophy in CA1 subfield and the microvascular lesion in the thalamus are both more severe in old apoE4/4 mice as compared with those in age-matched apoE3/3 mice. Doubts still exist on whether the decreased cell size in hippocampal CA1 subfield in old apoE4/4 mice is associated with dysfunction in learning and memory and whether the microvascular lesions indicate a higher risk of stroke in human apoE4 allele mice. To clarify these issues, further investigations are needed.


Assuntos
Transtornos Cerebrovasculares/genética , Transtornos Cerebrovasculares/patologia , Hipocampo/patologia , Células Piramidais/patologia , Tálamo/patologia , Fatores Etários , Análise de Variância , Animais , Apolipoproteína E3/deficiência , Apolipoproteína E3/genética , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Peso Corporal/genética , Feminino , Proteína Glial Fibrilar Ácida , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Tamanho do Órgão/genética , Células Piramidais/metabolismo
17.
Neural Plast ; 2007: 91236, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17710250

RESUMO

Anxiety is most common among Alzheimer's disease (AD) patients with an age at onset under age 65. Apolipoprotein E4 (apoE4) is a risk factor for developing AD at an earlier age and might contribute to this effect. In mice, apoE plays a role in the regulation of anxiety, which might involve histamine receptor-mediated signaling and steroidogenesis in the adrenal gland. In addition, human apoE isoforms have differential effects on anxiety in adult mice lacking apoE and probable AD patients. Compared to wild-type mice, mice lacking apoE and apoE4 mice showed pathological alterations in the central nucleus of the amygdala, which is involved in regulation of anxiety. ApoE4, but not mice lacking apoE, or apoE3 mice showed impaired dexamethasone suppression of plasma corticosterone. Understanding how apoE modulates measures of anxiety might help the developments of therapeutic targets to reduce or even prevent measures of anxiety in health and in dementing illnesses.


Assuntos
Ansiedade/metabolismo , Apolipoproteína E3/fisiologia , Apolipoproteína E4/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Tonsila do Cerebelo/fisiopatologia , Animais , Ansiedade/genética , Ansiedade/patologia , Ansiedade/psicologia , Apolipoproteína E3/deficiência , Apolipoproteína E3/genética , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Humanos , Camundongos
18.
Behav Brain Res ; 176(2): 372-5, 2007 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-17126420

RESUMO

The human apolipoprotein E isoforms, apoE2, apoE3, and apoE4, have differential effects on brain function. Compared to apoE3, apoE4 increases the risk of age-related cognitive decline in humans and female mice expressing apoE in neurons. Here, we show impaired spatial memory retention in female mice expressing apoE4 in astrocytes compared to those expressing apoE3 in astrocytes or lacking apoE. Thus, apoE4 impairs cognition whether expressed in neurons or astrocytes.


Assuntos
Apolipoproteína E4/fisiologia , Proteína Glial Fibrilar Ácida/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/patologia , Comportamento Espacial/fisiologia , Análise de Variância , Animais , Apolipoproteína E4/deficiência , Astrócitos/metabolismo , Comportamento Animal , Encéfalo/patologia , Feminino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Tempo de Reação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA