Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 142(6): 857-67, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20817278

RESUMO

Alzheimer's Disease (AD) is complicated by pro-oxidant intraneuronal Fe(2+) elevation as well as extracellular Zn(2+) accumulation within amyloid plaque. We found that the AD ß-amyloid protein precursor (APP) possesses ferroxidase activity mediated by a conserved H-ferritin-like active site, which is inhibited specifically by Zn(2+). Like ceruloplasmin, APP catalytically oxidizes Fe(2+), loads Fe(3+) into transferrin, and has a major interaction with ferroportin in HEK293T cells (that lack ceruloplasmin) and in human cortical tissue. Ablation of APP in HEK293T cells and primary neurons induces marked iron retention, whereas increasing APP695 promotes iron export. Unlike normal mice, APP(-/-) mice are vulnerable to dietary iron exposure, which causes Fe(2+) accumulation and oxidative stress in cortical neurons. Paralleling iron accumulation, APP ferroxidase activity in AD postmortem neocortex is inhibited by endogenous Zn(2+), which we demonstrate can originate from Zn(2+)-laden amyloid aggregates and correlates with Aß burden. Abnormal exchange of cortical zinc may link amyloid pathology with neuronal iron accumulation in AD.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Ceruloplasmina/antagonistas & inibidores , Zinco/metabolismo , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Animais , Linhagem Celular , Ceruloplasmina/química , Ceruloplasmina/metabolismo , Humanos , Ferro/metabolismo , Camundongos , Alinhamento de Sequência
2.
Exp Eye Res ; 221: 108974, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35202705

RESUMO

Neuropathological hallmarks of Alzheimer's disease (AD) include pathogenic accumulation of amyloid-ß (Aß) peptides and age-dependent formation of amyloid plaques in the brain. AD-associated Aß neuropathology begins decades before onset of cognitive symptoms and slowly progresses over the course of the disease. We previously reported discovery of Aß deposition, ß-amyloidopathy, and co-localizing supranuclear cataracts (SNC) in lenses from people with AD, but not other neurodegenerative disorders or normal aging. We confirmed AD-associated Aß molecular pathology in the lens by immunohistopathology, amyloid histochemistry, immunoblot analysis, epitope mapping, immunogold electron microscopy, quantitative immunoassays, and tryptic digest mass spectrometry peptide sequencing. Ultrastructural analysis revealed that AD-associated Aß deposits in AD lenses localize as electron-dense microaggregates in the cytoplasm of supranuclear (deep cortex) fiber cells. These Aß microaggregates also contain αB-crystallin and scatter light, thus linking Aß pathology and SNC phenotype expression in the lenses of people with AD. Subsequent research identified Aß lens pathology as the molecular origin of the distinctive cataracts associated with Down syndrome (DS, trisomy 21), a chromosomal disorder invariantly associated with early-onset Aß accumulation and Aß amyloidopathy in the brain. Investigation of 1249 participants in the Framingham Eye Study found that AD-associated quantitative traits in brain and lens are co-heritable. Moreover, AD-associated lens traits preceded MRI brain traits and cognitive deficits by a decade or more and predicted future AD. A genome-wide association study of bivariate outcomes in the same subjects identified a new AD risk factor locus in the CTNND2 gene encoding δ-catenin, a protein that modulates Aß production in brain and lens. Here we report identification of AD-related human Aß (hAß) lens pathology and age-dependent SNC phenotype expression in the Tg2576 transgenic mouse model of AD. Tg2576 mice express Swedish mutant human amyloid precursor protein (APP-Swe), accumulate hAß peptides and amyloid pathology in the brain, and exhibit cognitive deficits that slowly progress with increasing age. We found that Tg2576 trangenic (Tg+) mice, but not non-transgenic (Tg-) control mice, also express human APP, accumulate hAß peptides, and develop hAß molecular and ultrastructural pathologies in the lens. Tg2576 Tg+ mice exhibit age-dependent Aß supranuclear lens opacification that recapitulates lens pathology and SNC phenotype expression in human AD. In addition, we detected hAß in conditioned medium from lens explant cultures prepared from Tg+ mice, but not Tg- control mice, a finding consistent with constitutive hAß generation in the lens. In vitro studies showed that hAß promoted mouse lens protein aggregation detected by quasi-elastic light scattering (QLS) spectroscopy. These results support mechanistic (genotype-phenotype) linkage between Aß pathology and AD-related phenotypes in lens and brain. Collectively, our findings identify Aß pathology as the shared molecular etiology of two age-dependent AD-related cataracts associated with two human diseases (AD, DS) and homologous murine cataracts in the Tg2576 transgenic mouse model of AD. These results represent the first evidence of AD-related Aß pathology outside the brain and point to lens Aß as an optically-accessible AD biomarker for early detection and longitudinal monitoring of this devastating neurodegenerative disease.


Assuntos
Doença de Alzheimer , Catarata , Doenças Neurodegenerativas , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/patologia , Catarata/patologia , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Camundongos Transgênicos , Doenças Neurodegenerativas/patologia
3.
Proc Natl Acad Sci U S A ; 112(31): 9734-9, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26199414

RESUMO

Near-infrared fluorescence (NIRF) molecular imaging has been widely applied to monitoring therapy of cancer and other diseases in preclinical studies; however, this technology has not been applied successfully to monitoring therapy for Alzheimer's disease (AD). Although several NIRF probes for detecting amyloid beta (Aß) species of AD have been reported, none of these probes has been used to monitor changes of Aßs during therapy. In this article, we demonstrated that CRANAD-3, a curcumin analog, is capable of detecting both soluble and insoluble Aß species. In vivo imaging showed that the NIRF signal of CRANAD-3 from 4-mo-old transgenic AD (APP/PS1) mice was 2.29-fold higher than that from age-matched wild-type mice, indicating that CRANAD-3 is capable of detecting early molecular pathology. To verify the feasibility of CRANAD-3 for monitoring therapy, we first used the fast Aß-lowering drug LY2811376, a well-characterized beta-amyloid cleaving enzyme-1 inhibitor, to treat APP/PS1 mice. Imaging data suggested that CRANAD-3 could monitor the decrease in Aßs after drug treatment. To validate the imaging capacity of CRANAD-3 further, we used it to monitor the therapeutic effect of CRANAD-17, a curcumin analog for inhibition of Aß cross-linking. The imaging data indicated that the fluorescence signal in the CRANAD-17-treated group was significantly lower than that in the control group, and the result correlated with ELISA analysis of brain extraction and Aß plaque counting. It was the first time, to our knowledge, that NIRF was used to monitor AD therapy, and we believe that our imaging technology has the potential to have a high impact on AD drug development.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Monitoramento de Medicamentos , Imagem Molecular/métodos , Espectroscopia de Luz Próxima ao Infravermelho , Animais , Benzotiazóis , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Modelos Animais de Doenças , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Humanos , Camundongos Transgênicos , Fótons , Presenilina-1/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Espectrometria de Fluorescência , Tiazóis/metabolismo , Extratos de Tecidos , Titulometria
4.
Alzheimers Dement ; 14(12): 1602-1614, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30314800

RESUMO

OBJECTIVE: We explore here a novel model for amyloidogenesis in Alzheimer's disease (AD). This new perspective on AD amyloidosis seeks to provide a rational framework for incorporating recent and seemingly independent findings on the antimicrobial role of ß-amyloid and emerging experimental, genetic, and epidemiological data, suggesting innate immune-mediated inflammation propagates AD neurodegeneration. BACKGROUND: AD pathology is characterized by cerebral deposition of amyloid-ß protein (Aß) as ß-amyloid. Genetic studies have confirmed the key role of Aß in AD, revealing that mutation-mediated shifts in the peptides generation lead to early onset familial Alzheimer's disease. However, Aß generation appears normal for the majority of AD patients, who lack early onset familial Alzheimer's disease mutations. In prevailing models of nonfamilial AD, individual genetics and age-associated changes in brain milieu promote an intrinsically abnormal propensity of Aß for self-association. However, emerging findings are increasingly inconsistent with characterization of Aß oligomerization as a nonphysiological and exclusively pathological activity. Recent studies suggest Aß is an ancient, highly conserved effector molecule of innate immunity. Moreover, Aß oligomerization and ß-amyloid generation appear to be important innate immune pathways that mediate pathogen entrapment and protect against infection. NEW AD AMYLOIDOGENESIS MODEL: Recent findings on inflammation-mediated neurodegeneration and the role of Aß in immunity have led to emergence of the "Antimicrobial Protection Hypothesis" of AD. In this model, ß-amyloid deposition is an early innate immune response to genuine, or mistakenly perceived, immunochallenge. Aß first entraps and neutralizes invading pathogens in ß-amyloid. Aß fibrillization drives neuroinflammatory pathways that help fight the infection and clear ß-amyloid/pathogen deposits. In AD, chronic activation of this pathway leads to sustained inflammation and neurodegeneration. Mounting data link elevated brain microbe levels with AD. The Antimicrobial Protection Hypothesis reveals how increased brain microbial burden may directly exacerbate ß-amyloid deposition, inflammation, and AD progression. AMYLOID CASCADE HYPOTHESIS: In the antimicrobial protection model, the modality of Aß's pathophysiology is shifted from abnormal stochastic behavior toward dysregulated innate immune response. However, ß-amyloid deposition in AD still leads to neurodegeneration. Thus, the new model extends but remains broadly consistent with the Amyloid Cascade Hypothesis and overwhelming data showing the primacy of Aß in AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Anti-Infecciosos/metabolismo , Amiloidose/metabolismo , Animais , Humanos , Imunidade Inata/fisiologia , Modelos Biológicos , Neuroproteção/fisiologia
5.
Hum Mol Genet ; 18(20): 3987-96, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19608551

RESUMO

ADAM10, a member of a disintegrin and metalloprotease family, is an alpha-secretase capable of anti-amyloidogenic proteolysis of the amyloid precursor protein. Here, we present evidence for genetic association of ADAM10 with Alzheimer's disease (AD) as well as two rare potentially disease-associated non-synonymous mutations, Q170H and R181G, in the ADAM10 prodomain. These mutations were found in 11 of 16 affected individuals (average onset age 69.5 years) from seven late-onset AD families. Each mutation was also found in one unaffected subject implying incomplete penetrance. Functionally, both mutations significantly attenuated alpha-secretase activity of ADAM10 (>70% decrease), and elevated Abeta levels (1.5-3.5-fold) in cell-based studies. In summary, we provide the first evidence of ADAM10 as a candidate AD susceptibility gene, and report two potentially pathogenic mutations with incomplete penetrance for late-onset familial AD.


Assuntos
Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Proteína ADAM10 , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Polimorfismo de Nucleotídeo Único
6.
J Gerontol A Biol Sci Med Sci ; 75(9): e53-e62, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32515825

RESUMO

The absence of clinical tools to evaluate individual variation in the pace of aging represents a major impediment to understanding aging and maximizing health throughout life. The human lens is an ideal tissue for quantitative assessment of molecular aging in vivo. Long-lived proteins in lens fiber cells are expressed during fetal life, do not undergo turnover, accumulate molecular alterations throughout life, and are optically accessible in vivo. We used quasi-elastic light scattering (QLS) to measure age-dependent signals in lenses of healthy human subjects. Age-dependent QLS signal changes detected in vivo recapitulated time-dependent changes in hydrodynamic radius, protein polydispersity, and supramolecular order of human lens proteins during long-term incubation (~1 year) and in response to sustained oxidation (~2.5 months) in vitro. Our findings demonstrate that QLS analysis of human lens proteins provides a practical technique for noninvasive assessment of molecular aging in vivo.


Assuntos
Envelhecimento/fisiologia , Cristalinas/fisiologia , Difusão Dinâmica da Luz , Cristalino/fisiologia , Adolescente , Adulto , Criança , Pré-Escolar , Estudos Transversais , Cristalinas/química , Difusão Dinâmica da Luz/métodos , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Oxirredução , Adulto Jovem
7.
Ann Neurol ; 64(6): 618-27, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19006075

RESUMO

OBJECTIVE: An estimated 200 million patients worldwide have surgery each year. Anesthesia and surgery have been reported to facilitate emergence of Alzheimer's disease. The commonly used inhalation anesthetic isoflurane has previously been reported to induce apoptosis, and to increase levels and aggregation of Alzheimer's disease-associated amyloid beta-protein (Abeta) in cultured cells. However, the in vivo relevance has not been addressed. METHODS: We therefore set out to determine effects of isoflurane on caspase activation and levels of beta-site amyloid precursor protein-cleaving enzyme (BACE) and Abeta in naive mice, using Western blot, immunohistochemistry, and reverse transcriptase polymerase chain reaction. RESULTS: Here we show for the first time that a clinically relevant isoflurane anesthesia (1.4% isoflurane for 2 hours) leads to caspase activation and modest increases in levels of BACE 6 hours after anesthesia in mouse brain. Isoflurane anesthesia induces caspase activation, and increases levels of BACE and Abeta up to 24 hours after anesthesia. Isoflurane may increase BACE levels by reducing BACE degradation. Moreover, the Abeta aggregation inhibitor, clioquinol, was able to attenuate isoflurane-induced caspase-3 activation in vivo. INTERPRETATION: Given that transient insults to brain may lead to long-term brain damage, these findings suggest that isoflurane may promote Alzheimer's disease neuropathogenesis and, as such, have implications for use of isoflurane in humans, pending human study confirmation.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Anestésicos Inalatórios/toxicidade , Caspases/metabolismo , Isoflurano/toxicidade , Peptídeos beta-Amiloides/análise , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Caspases/análise , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
8.
Front Aging Neurosci ; 11: 70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30983989

RESUMO

The argument is frequently made that the amyloid-ß protein (Aß) persists in the human genome because Alzheimer's disease (AD) primarily afflicts individuals over reproductive age and, therefore, there is low selective pressure for the peptide's elimination or modification. This argument is an important premise for AD amyloidosis models and therapeutic strategies that characterize Aß as a functionless and intrinsically pathological protein. Here, we review if evolutionary theory and data on the genetics and biology of Aß are consistent with low selective pressure for the peptide's expression in senescence. Aß is an ancient neuropeptide expressed across vertebrates. Consistent with unusually high evolutionary selection constraint, the human Aß sequence is shared by a majority of vertebrate species and has been conserved across at least 400 million years. Unlike humans, the overwhelming majority of vertebrate species do not cease reproduction in senescence and selection pressure is maintained into old age. Hence, low selective pressure in senescence does not explain the persistence of Aß across the vertebrate genome. The "Grandmother hypothesis" (GMH) is the prevailing model explaining the unusual extended postfertile period of humans. In the GMH, high risk associated with birthing in old age has lead to early cessation of reproduction and a shift to intergenerational care of descendants. The rechanneling of resources to grandchildren by postreproductive individuals increases reproductive success of descendants. In the GMH model, selection pressure does not end following menopause. Thus, evolutionary models and phylogenetic data are not consistent with the absence of reproductive selection pressure for Aß among aged vertebrates, including humans. Our analysis suggests an alternative evolutionary model for the persistence of Aß in the vertebrate genome. Aß has recently been identified as an antimicrobial effector molecule of innate immunity. High conservation across the Chordata phylum is consistent with strong positive selection pressure driving human Aß's remarkable evolutionary longevity. Ancient origins and widespread conservation suggest the human Aß sequence is highly optimized for its immune role. We detail our analysis and discuss how the emerging "Antimicrobial Protection Hypothesis" of AD may provide insights into possible evolutionary roles for Aß in infection, aging, and disease etiology.

9.
Neuron ; 44(2): 227-38, 2004 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-15473963

RESUMO

Amyloid beta-peptide (Abeta) accumulation in specific brain regions is a pathological hallmark of Alzheimer's disease (AD). We have previously reported that a well-characterized acyl-coenzyme A: cholesterol acyltransferase (ACAT) inhibitor, CP-113,818, inhibits Abeta production in cell-based experiments. Here, we assessed the efficacy of CP-113,818 in reducing AD-like pathology in the brains of transgenic mice expressing human APP(751) containing the London (V717I) and Swedish (K670M/N671L) mutations. Two months of treatment with CP-113,818 reduced the accumulation of amyloid plaques by 88%-99% and membrane/insoluble Abeta levels by 83%-96%, while also decreasing brain cholesteryl-esters by 86%. Additionally, soluble Abeta(42) was reduced by 34% in brain homogenates. Spatial learning was slightly improved and correlated with decreased Abeta levels. In nontransgenic littermates, CP-113,818 also reduced ectodomain shedding of endogenous APP in the brain. Our results suggest that ACAT inhibition may be effective in the prevention and treatment of AD by inhibiting generation of the Abeta peptide.


Assuntos
Peptídeos beta-Amiloides/efeitos dos fármacos , Encéfalo/patologia , Inibidores Enzimáticos/uso terapêutico , Piridinas/uso terapêutico , Esterol O-Aciltransferase/efeitos dos fármacos , Glândulas Suprarrenais/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Ésteres do Colesterol/análise , Ésteres do Colesterol/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/efeitos adversos , Feminino , Humanos , Aprendizagem/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo , Piridinas/efeitos adversos , Fatores Sexuais , Esterol O-Aciltransferase/metabolismo
10.
J Neurosci ; 27(6): 1247-54, 2007 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-17287498

RESUMO

The anesthetic isoflurane has been reported to induce apoptosis and increase Abeta generation and aggregation. However, the molecular mechanism underlying these effects remains unknown. We therefore set out to assess whether the effects of isoflurane on apoptosis are linked to amyloid beta-protein (Abeta) generation and aggregation. For this purpose, we assessed the effects of isoflurane on beta-site amyloid beta precursor protein (APP)-cleaving enzyme (BACE) and gamma-secretase, the proteases responsible for Abeta generation. We also tested the effects of inhibitors of Abeta aggregation (iAbeta5, a beta-sheet breaker peptide; clioquinol, a copper-zinc chelator) on the ability of isoflurane to induce apoptosis. All of these studies were performed on naive human H4 neuroglioma cells as well as those overexpressing APP (H4-APP cells). Isoflurane increased the levels of BACE and gamma-secretase and secreted Abeta in the H4-APP cells. Isoflurane-induced Abeta generation could be blocked by the broad-based caspase inhibitor Z-VAD. The Abeta aggregation inhibitors, iAbeta5 and clioquinol, selectively attenuated caspase-3 activation induced by isoflurane. However, isoflurane was able to induce caspase-3 activation in the absence of any detectable alterations of Abeta generation in naive H4 cells. Finally, Abeta potentiated the isoflurane-induced caspase-3 activation in naive H4 cells. Collectively, these findings suggest that isoflurane can induce apoptosis, which, in turn, increases BACE and gamma-secretase levels and Abeta secretion. Isoflurane also promotes Abeta aggregation. Accumulation of aggregated Abeta in the media can then promote apoptosis. The result is a vicious cycle of isoflurane-induced apoptosis, Abeta generation and aggregation, and additional rounds of apoptosis, leading to cell death.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anestésicos Inalatórios/toxicidade , Apoptose/efeitos dos fármacos , Isoflurano/toxicidade , Neuroglia/efeitos dos fármacos , Doença de Alzheimer/induzido quimicamente , Secretases da Proteína Precursora do Amiloide/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Anestésicos Inalatórios/efeitos adversos , Anestésicos Inalatórios/farmacologia , Ácido Aspártico Endopeptidases/efeitos dos fármacos , Ácido Aspártico Endopeptidases/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Quelantes/farmacologia , Clioquinol/farmacologia , Cobre , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ganglioglioma/patologia , Humanos , Isoflurano/efeitos adversos , Isoflurano/farmacologia , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Zinco
11.
Neuron ; 99(1): 56-63.e3, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-30001512

RESUMO

Amyloid-ß peptide (Aß) fibrilization and deposition as ß-amyloid are hallmarks of Alzheimer's disease (AD) pathology. We recently reported Aß is an innate immune protein that protects against fungal and bacterial infections. Fibrilization pathways mediate Aß antimicrobial activities. Thus, infection can seed and dramatically accelerate ß-amyloid deposition. Here, we show Aß oligomers bind herpesvirus surface glycoproteins, accelerating ß-amyloid deposition and leading to protective viral entrapment activity in 5XFAD mouse and 3D human neural cell culture infection models against neurotropic herpes simplex virus 1 (HSV1) and human herpesvirus 6A and B. Herpesviridae are linked to AD, but it has been unclear how viruses may induce ß-amyloidosis in brain. These data support the notion that Aß might play a protective role in CNS innate immunity, and suggest an AD etiological mechanism in which herpesviridae infection may directly promote Aß amyloidosis.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Encéfalo/metabolismo , Encefalite Viral/metabolismo , Herpesviridae , Doença de Alzheimer/virologia , Amiloidose/virologia , Animais , Encéfalo/virologia , Células Cultivadas , Modelos Animais de Doenças , Encefalite por Herpes Simples/metabolismo , Encefalite por Herpes Simples/virologia , Encefalite Viral/virologia , Herpesvirus Humano 1 , Herpesvirus Humano 6 , Humanos , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Neurônios , Placa Amiloide/metabolismo , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/virologia
12.
Sci Transl Med ; 8(340): 340ra72, 2016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27225182

RESUMO

The amyloid-ß peptide (Aß) is a key protein in Alzheimer's disease (AD) pathology. We previously reported in vitro evidence suggesting that Aß is an antimicrobial peptide. We present in vivo data showing that Aß expression protects against fungal and bacterial infections in mouse, nematode, and cell culture models of AD. We show that Aß oligomerization, a behavior traditionally viewed as intrinsically pathological, may be necessary for the antimicrobial activities of the peptide. Collectively, our data are consistent with a model in which soluble Aß oligomers first bind to microbial cell wall carbohydrates via a heparin-binding domain. Developing protofibrils inhibited pathogen adhesion to host cells. Propagating ß-amyloid fibrils mediate agglutination and eventual entrapment of unatttached microbes. Consistent with our model, Salmonella Typhimurium bacterial infection of the brains of transgenic 5XFAD mice resulted in rapid seeding and accelerated ß-amyloid deposition, which closely colocalized with the invading bacteria. Our findings raise the intriguing possibility that ß-amyloid may play a protective role in innate immunity and infectious or sterile inflammatory stimuli may drive amyloidosis. These data suggest a dual protective/damaging role for Aß, as has been described for other antimicrobial peptides.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/microbiologia , Peptídeos beta-Amiloides/fisiologia , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Camundongos , Camundongos Transgênicos , Salmonella typhimurium/patogenicidade
13.
Curr Alzheimer Res ; 2(2): 269-73, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15974929

RESUMO

The pathogenesis of Alzheimer's disease (AD) involves the abnormal accumulation and deposition of beta-amyloid in cerebral blood vessels and in the brain parenchyma. Critical in modulating beta-amyloid deposition in brain is the flux of Abeta across the blood brain barrier. The low-density lipoprotein receptor-related protein (LRP), is a large endocytic receptor that mediates the efflux of Abeta out of brain and into the periphery. The first step in the LRP-mediated clearance of Abeta involves the formation of a complex between Abeta and the LRP ligands apolipoprotein E (apoE) or alpha(2)-macroglobulin (alpha(2)M). The Abeta/chaperone complexes then bind to LRP via binding sites on apoE or alpha(2)M. The efflux of Abeta/chaperone complexes out of the neuropil and into the periphery may be attenuated by LRP-ligands that compete with apoE or alpha(2)M for LRP binding. LRP is also the cell surface receptor for Kunitz Protease Inhibitor (KPI) containing isoforms of Abeta's parent protein, the amyloid protein precursor (APP). Protein and mRNA levels of KPI-containing APP isoforms (APP-KPI) are elevated in AD brain and are associated with increased Abeta production. In this study we show that soluble non-amyloidogenic APP-KPI can also inhibit the uptake of Abeta/alpha(2)M in a cell culture model of LRP mediated Abeta clearance. Clearance of Abeta/apoE complexes was not inhibited by APP-KPI. Our findings are consistent with studies showing that apoE and alpha(2)M have discrete binding sites on LRP. Most significantly, our data suggests that the elevated levels of APP-KPI in AD brain may attenuate the clearance of Abeta, the proteins own amyloidogenic catabolic product.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Aprotinina/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Ligação Proteica/fisiologia , Isoformas de Proteínas/metabolismo
14.
Exp Gerontol ; 39(11-12): 1641-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15582280

RESUMO

A growing body of evidence indicates that dysregulation of cerebral biometals (Fe, Cu, Zn) and their interactions with APP and Abeta amyloid may contribute to the Alzheimer's amyloid pathology, and thus metal chelation could be a rational therapeutic approach for interdicting AD pathogenesis. However, poor target specificity and consequential clinical safety of current metal-complexing agents have limited their widespread clinical use. To develop the next generation of metal chelators, we have designed and synthesized a new bifunctional molecule-XH1, based on a novel 'pharmacophore conjugation' concept. This lipophilic molecule has both amyloid-binding and metal-chelating moieties covalently connected by amide bonds. It achieved a putative binding geometry with Abeta1-40 peptide by the computational chemistry modeling and reduced Zn(II)-induced Abeta1-40 aggregation in vitro as determined by turbidometry. Moreover, our pilot data indicated that XH1 has no significant neurotoxicity at low micromolar concentrations and acute animal toxicity. XH1 specifically reduced APP protein expression in human SH-SY5Y neuroblastoma cells and attenuated cerebral Abeta amyloid pathology in PS1/APP transgenic mice without inducing apparent toxicity and behavior disturbances. Collectively, these preliminary findings carry implication for XH1 being a BBB-permeable lead compound for AD therapeutics targeting Alzheimer's amyloidogenesis, although further studies are needed.


Assuntos
Doença de Alzheimer/prevenção & controle , Amiloidose/tratamento farmacológico , Quelantes/uso terapêutico , Metais , Idoso , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/análise , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Animais , Encéfalo/metabolismo , Química Encefálica , Neoplasias Encefálicas , Linhagem Celular Tumoral , Quelantes/síntese química , Feminino , Humanos , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Neuroblastoma , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo
15.
Ann N Y Acad Sci ; 1012: 153-63, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15105262

RESUMO

Considerable evidence is mounting that dyshomeostasis of the redox-active biometals, Cu and Fe, and oxidative stress contribute to the neuropathology of Alzheimer's disease (AD). Present data suggest that metals can interact directly with Abeta peptide, the principal component of beta-amyloid that is one of the primary lesions in AD. The binding of metals to Abeta modulates several physiochemical properties of Abeta that are thought to be central to the pathogenicity of the peptide. First, we and others have shown that metals can promote the in vitro aggregation into tinctorial Abeta amyloid. Studies have confirmed that insoluble amyloid plaques in postmortem AD brain are abnormally enriched in Cu, Fe, and Zn. Conversely, metal chelators dissolve these proteinaceous deposits from postmortem AD brain tissue and attenuate cerebral Abeta amyloid burden in APP transgenic mouse models of AD. Second, we have demonstrated that redox-active Cu(II) and, to a lesser extent, Fe(III) are reduced in the presence of Abeta with concomitant production of reactive oxygen species (ROS), hydrogen peroxide (H(2)O(2)) and hydroxyl radical (OH*). These Abeta/metal redox reactions, which are silenced by redox-inert Zn(II), but exacerbated by biological reducing agents, may lead directly to the widespread oxidation damages observed in AD brains. Moreover, studies have also shown that H(2)O(2) mediates Abeta cellular toxicity and increases the production of both Abeta and amyloid precursor protein (APP). Third, the 5' untranslated region (5'UTR) of APP mRNA has a functional iron-response element (IRE), which is consistent with biochemical evidence that APP is a redox-active metalloprotein. Hence, the redox interactions between Abeta, APP, and metals may be at the heart of a pathological positive feedback system wherein Abeta amyloidosis and oxidative stress promote each other. The emergence of redox-active metals as key players in AD pathogenesis strongly argues that amyloid-specific metal-complexing agents and antioxidants be investigated as possible disease-modifying agents for treating this horrible disease.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Metais/metabolismo , Estresse Oxidativo , Doença de Alzheimer/tratamento farmacológico , Amiloide/metabolismo , Animais , Antioxidantes/uso terapêutico , Quelantes/uso terapêutico , Humanos , Oxirredução
16.
Dis Model Mech ; 7(3): 373-85, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24487408

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory loss and decreased synaptic function. Advances in transgenic animal models of AD have facilitated our understanding of this disorder, and have aided in the development, speed and efficiency of testing potential therapeutics. Recently, we have described the characterization of a novel model of AD in the fruit fly, Drosophila melanogaster, where we expressed the human AD-associated proteins APP and BACE in the central nervous system of the fly. Here we describe synaptic defects in the larval neuromuscular junction (NMJ) in this model. Our results indicate that expression of human APP and BACE at the larval NMJ leads to defective larval locomotion behavior, decreased presynaptic connections, altered mitochondrial localization in presynaptic motor neurons and decreased postsynaptic protein levels. Treating larvae expressing APP and BACE with the γ-secretase inhibitor L-685,458 suppresses the behavioral defects as well as the pre- and postsynaptic defects. We suggest that this model will be useful to assess and model the synaptic dysfunction normally associated with AD, and will also serve as a powerful in vivo tool for rapid testing of potential therapeutics for AD.


Assuntos
Doença de Alzheimer/patologia , Drosophila melanogaster/fisiologia , Sinapses/patologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Ácido Aspártico Endopeptidases/metabolismo , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Inibidores Enzimáticos/farmacologia , Humanos , Larva/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Fenótipo , Transporte Proteico/efeitos dos fármacos , Sinapses/metabolismo , Transgenes
19.
Sci Transl Med ; 4(134): 134ra60, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22593173

RESUMO

Blast exposure is associated with traumatic brain injury (TBI), neuropsychiatric symptoms, and long-term cognitive disability. We examined a case series of postmortem brains from U.S. military veterans exposed to blast and/or concussive injury. We found evidence of chronic traumatic encephalopathy (CTE), a tau protein-linked neurodegenerative disease, that was similar to the CTE neuropathology observed in young amateur American football players and a professional wrestler with histories of concussive injuries. We developed a blast neurotrauma mouse model that recapitulated CTE-linked neuropathology in wild-type C57BL/6 mice 2 weeks after exposure to a single blast. Blast-exposed mice demonstrated phosphorylated tauopathy, myelinated axonopathy, microvasculopathy, chronic neuroinflammation, and neurodegeneration in the absence of macroscopic tissue damage or hemorrhage. Blast exposure induced persistent hippocampal-dependent learning and memory deficits that persisted for at least 1 month and correlated with impaired axonal conduction and defective activity-dependent long-term potentiation of synaptic transmission. Intracerebral pressure recordings demonstrated that shock waves traversed the mouse brain with minimal change and without thoracic contributions. Kinematic analysis revealed blast-induced head oscillation at accelerations sufficient to cause brain injury. Head immobilization during blast exposure prevented blast-induced learning and memory deficits. The contribution of blast wind to injurious head acceleration may be a primary injury mechanism leading to blast-related TBI and CTE. These results identify common pathogenic determinants leading to CTE in blast-exposed military veterans and head-injured athletes and additionally provide mechanistic evidence linking blast exposure to persistent impairments in neurophysiological function, learning, and memory.


Assuntos
Traumatismos por Explosões/complicações , Traumatismos por Explosões/patologia , Lesão Encefálica Crônica/complicações , Lesão Encefálica Crônica/patologia , Militares/psicologia , Veteranos/psicologia , Aceleração , Adolescente , Adulto , Animais , Atletas , Axônios/patologia , Comportamento Animal , Traumatismos por Explosões/fisiopatologia , Concussão Encefálica/complicações , Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Lesão Encefálica Crônica/fisiopatologia , Modelos Animais de Doenças , Cabeça/patologia , Cabeça/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Humanos , Pressão Intracraniana , Potenciação de Longa Duração , Masculino , Camundongos , Pessoa de Meia-Idade , Fosforilação , Mudanças Depois da Morte , Transmissão Sináptica , Adulto Jovem , Proteínas tau/metabolismo
20.
PLoS One ; 6(4): e19362, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559413

RESUMO

BACKGROUND: Systematic differentiation of amyloid (Aß) species could be important for diagnosis of Alzheimer's disease (AD). In spite of significant progress, controversies remain regarding which species are the primary contributors to the AD pathology, and which species could be used as the best biomarkers for its diagnosis. These controversies are partially caused by the lack of reliable methods to differentiate the complicated subtypes of Aß species. Particularly, differentiation of Aß monomers from toxic higher molecular weight species (HrMW) would be beneficial for drug screening, diagnosis, and molecular mechanism studies. However, fast and cheap methods for these specific aims are still lacking. PRINCIPAL FINDINGS: We demonstrated the feasibility of a non-conjugated FRET (Förster resonance energy transfer) technique that utilized amyloid beta (Aß) species as intrinsic platforms for the FRET pair assembly. Mixing two structurally similar curcumin derivatives that served as the small molecule FRET pair with Aß40 aggregates resulted in a FRET signal, while no signal was detected when using Aß40 monomer solution. Lastly, this FRET technique enabled us to quantify the concentrations of Aß monomers and high molecular weight species in solution. SIGNIFICANCE: We believe that this FRET technique could potentially be used as a tool for screening for inhibitors of Aß aggregation. We also suggest that this concept could be generalized to other misfolded proteins/peptides implicated in various pathologies including amyloid in diabetes, prion in bovine spongiform encephalopathy, tau protein in AD, and α-synuclein in Parkinson disease.


Assuntos
Peptídeos beta-Amiloides/química , Transferência Ressonante de Energia de Fluorescência/métodos , Peptídeos beta-Amiloides/isolamento & purificação , Bioquímica/métodos , Curcumina/química , Dimerização , Humanos , Peso Molecular , Fragmentos de Peptídeos/química , Ligação Proteica , Desnaturação Proteica , Dobramento de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA