Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
2.
Proc Natl Acad Sci U S A ; 120(41): e2300258120, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37801475

RESUMO

Despite much effort, antibody therapies for Alzheimer's disease (AD) have shown limited efficacy. Challenges to the rational design of effective antibodies include the difficulty of achieving specific affinity to critical targets, poor expression, and antibody aggregation caused by buried charges and unstructured loops. To overcome these challenges, we grafted previously determined sequences of fibril-capping amyloid inhibitors onto a camel heavy chain antibody scaffold. These sequences were designed to cap fibrils of tau, known to form the neurofibrillary tangles of AD, thereby preventing fibril elongation. The nanobodies grafted with capping inhibitors blocked tau aggregation in biosensor cells seeded with postmortem brain extracts from AD and progressive supranuclear palsy (PSP) patients. The tau capping nanobody inhibitors also blocked seeding by recombinant tau oligomers. Another challenge to the design of effective antibodies is their poor blood-brain barrier (BBB) penetration. In this study, we also designed a bispecific nanobody composed of a nanobody that targets a receptor on the BBB and a tau capping nanobody inhibitor, conjoined by a flexible linker. We provide evidence that the bispecific nanobody improved BBB penetration over the tau capping inhibitor alone after intravenous administration in mice. Our results suggest that the design of synthetic antibodies that target sequences that drive protein aggregation may be a promising approach to inhibit the prion-like seeding of tau and other proteins involved in AD and related proteinopathies.


Assuntos
Doença de Alzheimer , Anticorpos de Domínio Único , Paralisia Supranuclear Progressiva , Humanos , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Proteínas tau/metabolismo , Anticorpos de Domínio Único/farmacologia , Anticorpos de Domínio Único/metabolismo , Emaranhados Neurofibrilares/metabolismo , Paralisia Supranuclear Progressiva/metabolismo , Anticorpos/metabolismo , Encéfalo/metabolismo
3.
Alzheimers Dement ; 19(6): 2239-2252, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36448627

RESUMO

INTRODUCTION: The inositol polyphosphate-5-phosphatase D (INPP5D) gene encodes a dual-specificity phosphatase that can dephosphorylate both phospholipids and phosphoproteins. Single nucleotide polymorphisms in INPP5D impact risk for developing late onset sporadic Alzheimer's disease (LOAD). METHODS: To assess the consequences of inducible Inpp5d knockdown in microglia of APPKM670/671NL /PSEN1Δexon9 (PSAPP) mice, we injected 3-month-old Inpp5dfl/fl /Cx3cr1CreER/+ and PSAPP/Inpp5dfl/fl /Cx3cr1CreER/+ mice with either tamoxifen (TAM) or corn oil (CO) to induce recombination. RESULTS: At age 6 months, we found that the percent area of 6E10+ deposits and plaque-associated microglia in Inpp5d knockdown mice were increased compared to controls. Spatial transcriptomics identified a plaque-specific expression profile that was extensively altered by Inpp5d knockdown. DISCUSSION: These results demonstrate that conditional Inpp5d downregulation in the PSAPP mouse increases plaque burden and recruitment of microglia to plaques. Spatial transcriptomics highlighted an extended gene expression signature associated with plaques and identified CST7 (cystatin F) as a novel marker of plaques. HIGHLIGHTS: Inpp5d knockdown increases plaque burden and plaque-associated microglia number. Spatial transcriptomics identifies an expanded plaque-specific gene expression profile. Plaque-induced gene expression is altered by Inpp5d knockdown in microglia. Our plaque-associated gene signature overlaps with human Alzheimer's disease gene networks.


Assuntos
Doença de Alzheimer , Camundongos , Humanos , Animais , Lactente , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Microglia/metabolismo , Camundongos Transgênicos , Placa Amiloide/metabolismo , Modelos Animais de Doenças , Peptídeos beta-Amiloides/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/metabolismo
4.
J Alzheimers Dis ; 90(4): 1501-1521, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36278355

RESUMO

BACKGROUND: Many identified mechanisms could be upstream of the prominent amyloid-ß (Aß) plaques in Alzheimer's disease (AD). OBJECTIVE: To profile the progression of pathology in AD. METHODS: We monitored metabolic signaling, redox stress, intraneuronal amyloid-ß (iAß) accumulation, and extracellular plaque deposition in the brains of 3xTg-AD mice across the lifespan. RESULTS: Intracellular accumulation of aggregated Aß in the CA1 pyramidal cells at 9 months preceded extracellular plaques that first presented in the CA1 at 16 months of age. In biochemical assays, brain glutathione (GSH) declined with age in both 3xTg-AD and non-transgenic controls, but the decline was accelerated in 3xTg-AD brains from 2 to 4 months. The decline in GSH correlated exponentially with the rise in iAß. Integrated metabolic signaling as the ratio of phospho-Akt (pAkt) to total Akt (tAkt) in the PI3kinase and mTOR pathway declined at 6, 9, and 12 months, before rising at 16 and 20 months. These pAkt/tAkt ratios correlated with both iAß and GSH levels in a U-shaped relationship. Selective vulnerability of age-related AD-genotype-specific pAkt changes was greatest in the CA1 pyramidal cell layer. To demonstrate redox causation, iAß accumulation was lowered in cultured middle-age adult 3xTg-AD neurons by treatment of the oxidized redox state in the neurons with exogenous cysteine. CONCLUSION: The order of pathologic progression in the 3xTg-AD mouse was loss of GSH (oxidative redox shift) followed by a pAkt/tAkt metabolic shift in CA1, iAß accumulation in CA1, and extracellular Aß deposition. Upstream targets may prove strategically more effective for therapy before irreversible changes.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Camundongos Transgênicos , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/patologia , Modelos Animais de Doenças , Região CA1 Hipocampal/patologia , Glutationa/metabolismo
5.
Pregnancy Hypertens ; 28: 51-59, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35183929

RESUMO

Prior research has shown that urine of women with preeclampsia (PE) contains amyloid-like aggregates that are congophilic (exhibit affinity for the amyloidophilic dye Congo red) and immunoreactive with A11, a polyclonal serum against prefibrillar ß-amyloid oligomers, thereby supporting pathogenic similarity between PE and protein conformational disorders such as Alzheimer's and prion disease. The objective of this study was to interrogate PE urine using monoclonal antibodies with previously characterized A11-like epitopes. Over 100 conformation-dependent monoclonals were screened and three (mA11-09, mA11-89, and mA11-205) selected for further confirmation in 196 urine samples grouped as follows: severe features PE (sPE, n = 114), PE without severe features (mPE, n = 30), chronic hypertension (crHTN, n = 14) and normotensive pregnant control (P-CRL, n = 38). We showed that the selected conformation-specific monoclonals distinguished among patients with varying severities of PE from P-CRL and patients with crHTN. By use of latent class analysis (LCA) we identified three classes of subjects: Class 1 (n = 94) comprised patients whose urine was both congophilic and reactive with the monoclonals. These women were more likely diagnosed with early-onset sPE and had severe hypertension and proteinuria; Class 2 patients (n = 55) were negative for congophilia and against the antibodies. These were predominantly P-CRL and crHTN patients. Lastly, Class 3 patients (n = 48) were positive for urine congophilia, albeit at lower intensity, but negative for monoclonal immunoreactivities. These women were diagnosed primarily as mPE or late-onset sPE. Collectively, our study validates conformation-dependent Aß imunoreactivity of PE urine which in conjunction to urine congophilia may represent an additional indicator of disease severity.


Assuntos
Hipertensão , Pré-Eclâmpsia , Anticorpos Monoclonais , Vermelho Congo , Feminino , Humanos , Pré-Eclâmpsia/metabolismo , Gravidez , Proteinúria
6.
Arch Physiol Biochem ; 128(3): 732-739, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32046518

RESUMO

Alzheimer's disease (AD) is atrophy of brain cells that lead to decline in the mental capacity and memory. This study investigated the mechanism which postulates that intraneuronal accumulation of amyloid aggregates for pathogenesis of AD. The PC12 cell line was used to examine the amyloid beta (Aß) aggregation in different stages. It was found that dot-blot filter retardation assay for Ub-CTF was 0.25 and 0.2 µM for SS-CTF. In addition, incubation of SS-CTF with 200 µM Aß-42 then bounded with an antibody directed against Aß. It was suggested that most bound Aß-42 in the oligomeric form. Confocal microscope showed that stained with DAPI (blue) in the neuritic plaques, APP-GFP (green) and specific monoclonal M78 (red). Aß oligomeric taken up by neurons and accumulation of misfolded Aß aggregates continue in a perinuclear location. Fluorescence intensities correlate with the priming effect observed on the Aß (p < .001). It was concluded that a new amyloid hypothesis is promising in therapy development to reduce the incidence of disease by inhibition of intraneuronal amyloid aggregation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Neurônios/metabolismo , Células PC12 , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Ratos
7.
Arch Physiol Biochem ; 128(5): 1275-1282, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32449861

RESUMO

The current study identified the specific antibodies that recognise amyloid protein for Alzheimer disease - immunotherapy. The immune-selection of random sequences from a phage display library and sequencing to obtain the random 12 amino acids peptide library for each antibody, and then we analysed these peptides for unique and common sequences, relation to Aß42 sequence and shape and pattern of the amino acid reaction to the antibody to predict the epitopes. Data obtained for 4G8 showed that, the sequence segment related to the putative epitope of 4G8 was LVFFAED. Nine of the ten top sequences contain the sequence RHD corresponding to the Aß sequence from residues 5-7. Peptide 7 has the sequence IRYDTGSYHIH, which has a RYD. It was concluded that, 4G8 and 6E10 can tolerate the binding the sequences that explain it is able to recognise amyloid aggregates.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Aminoácidos , Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/genética , Anticorpos Monoclonais/metabolismo , Epitopos/química , Humanos , Imunoterapia , Biblioteca de Peptídeos
8.
J Biol Chem ; 295(31): 10662-10676, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32493775

RESUMO

Soluble oligomers of aggregated tau accompany the accumulation of insoluble amyloid fibrils, a histological hallmark of Alzheimer disease (AD) and two dozen related neurodegenerative diseases. Both oligomers and fibrils seed the spread of Tau pathology, and by virtue of their low molecular weight and relative solubility, oligomers may be particularly pernicious seeds. Here, we report the formation of in vitro tau oligomers formed by an ionic liquid (IL15). Using IL15-induced recombinant tau oligomers and a dot blot assay, we discovered a mAb (M204) that binds oligomeric tau, but not tau monomers or fibrils. M204 and an engineered single-chain variable fragment (scFv) inhibited seeding by IL15-induced tau oligomers and pathological extracts from donors with AD and chronic traumatic encephalopathy. This finding suggests that M204-scFv targets pathological structures that are formed by tau in neurodegenerative diseases. We found that M204-scFv itself partitions into oligomeric forms that inhibit seeding differently, and crystal structures of the M204-scFv monomer, dimer, and trimer revealed conformational differences that explain differences among these forms in binding and inhibition. The efficiency of M204-scFv antibodies to inhibit the seeding by brain tissue extracts from different donors with tauopathies varied among individuals, indicating the possible existence of distinct amyloid polymorphs. We propose that by binding to oligomers, which are hypothesized to be the earliest seeding-competent species, M204-scFv may have potential as an early-stage diagnostic for AD and tauopathies, and also could guide the development of promising therapeutic antibodies.


Assuntos
Doença de Alzheimer , Multimerização Proteica , Anticorpos de Cadeia Única/química , Proteínas tau/química , Cristalografia por Raios X , Humanos
9.
J Alzheimers Dis ; 73(1): 229-246, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31771065

RESUMO

This work provides new insight into the age-related basis of Alzheimer's disease (AD), the composition of intraneuronal amyloid (iAß), and the mechanism of an age-related increase in iAß in adult AD-model mouse neurons. A new end-specific antibody for Aß45 and another for aggregated forms of Aß provide new insight into the composition of iAß and the mechanism of accumulation in old adult neurons from the 3xTg-AD model mouse. iAß levels containing aggregates of Aß45 increased 30-50-fold in neurons from young to old age and were further stimulated upon glutamate treatment. iAß was 8 times more abundant in 3xTg-AD than non-transgenic neurons with imaged particle sizes following the same log-log distribution, suggesting a similar snow-ball mechanism of intracellular biogenesis. Pathologically misfolded and mislocalized Alz50 tau colocalized with iAß and rapidly increased following a brief metabolic stress with glutamate. AßPP-CTF, Aß45, and aggregated Aß colocalized most strongly with mitochondria and endosomes and less with lysosomes and autophagosomes. Differences in iAß by sex were minor. These results suggest that incomplete carboxyl-terminal trimming of long Aßs by gamma-secretase produced large intracellular deposits which limited completion of autophagy in aged neurons. Understanding the mechanism of age-related changes in iAß processing may lead to application of countermeasures to prolong dementia-free health span.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Autofagossomos/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Ácido Glutâmico/farmacologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Neurônios/ultraestrutura , Tamanho da Partícula
10.
Sci Rep ; 9(1): 15936, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31685865

RESUMO

Alzheimer's disease (AD), the most common age-related neurodegenerative disorder, is currently conceptualized as a disease of synaptic failure. Synaptic impairments are robust within the AD brain and better correlate with dementia severity when compared with other pathological features of the disease. Nevertheless, the series of events that promote synaptic failure still remain under debate, as potential triggers such as ß-amyloid (Aß) can vary in size, configuration and cellular location, challenging data interpretation in causation studies. Here we present data obtained using adeno-associated viral (AAV) constructs that drive the expression of oligomeric Aß either intra or extracellularly. We observed that expression of Aß in both cellular compartments affect learning and memory, reduce the number of synapses and the expression of synaptic-related proteins, and disrupt chemical long-term potentiation (cLTP). Together, these findings indicate that during the progression AD the early accumulation of Aß inside neurons is sufficient to promote morphological and functional cellular toxicity, a phenomenon that can be exacerbated by the buildup of Aß in the brain parenchyma. Moreover, our AAV constructs represent a valuable tool in the investigation of the pathological properties of Aß oligomers both in vivo and in vitro.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Dependovirus/genética , Hipocampo/metabolismo , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Células Cultivadas , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Hipocampo/citologia , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/genética , Sinapses/metabolismo
11.
Elife ; 82019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31612856

RESUMO

Alzheimer's disease (AD) pathology is characterized by plaques of amyloid beta (Aß) and neurofibrillary tangles of tau. Aß aggregation is thought to occur at early stages of the disease, and ultimately gives way to the formation of tau tangles which track with cognitive decline in humans. Here, we report the crystal structure of an Aß core segment determined by MicroED and in it, note characteristics of both fibrillar and oligomeric structure. Using this structure, we designed peptide-based inhibitors that reduce Aß aggregation and toxicity of already-aggregated species. Unexpectedly, we also found that these inhibitors reduce the efficiency of Aß-mediated tau aggregation, and moreover reduce aggregation and self-seeding of tau fibrils. The ability of these inhibitors to interfere with both Aß and tau seeds suggests these fibrils share a common epitope, and supports the hypothesis that cross-seeding is one mechanism by which amyloid is linked to tau aggregation and could promote cognitive decline.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Cristalografia por Raios X , Humanos , Estrutura Molecular , Ligação Proteica , Conformação Proteica
12.
Mol Neurodegener ; 13(1): 11, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29490706

RESUMO

BACKGROUND: Besides the two main classical features of amyloid beta aggregation and tau-containing neurofibrillary tangle deposition, neuroinflammation plays an important yet unclear role in the pathophysiology of Alzheimer's disease (AD). Microglia are believed to be key mediators of neuroinflammation during AD and responsible for the regulation of brain homeostasis by balancing neurotoxicity and neuroprotective events. We have previously reported evidence that neuritic plaques are derived from dead neurons that have accumulated intraneuronal amyloid and further recruit Iba1-positive cells, which play a role in either neuronal demise or neuritic plaque maturation or both. METHODS: To study the impact of microglia on neuritic plaque development, we treated two-month-old 5XFAD mice with a selective colony stimulation factor 1 receptor (CSF1R) inhibitor, PLX3397, for a period of 3 months, resulting in a significant ablation of microglia. Directly after this treatment, we analyzed the amount of intraneuronal amyloid and neuritic plaques and performed behavioral studies including Y-maze, fear conditioning and elevated plus maze. RESULTS: We found that early long-term PLX3397 administration results in a dramatic reduction of both intraneuronal amyloid as well as neuritic plaque deposition. PLX3397 treated young 5XFAD mice also displayed a significant decrease of soluble fibrillar amyloid oligomers in brain lysates, a depletion of soluble pre-fibrillar oligomers in plasma and an improvement in cognitive function measured by fear conditioning tests. CONCLUSIONS: Our findings demonstrate that CSF1R signaling, either directly on neurons or mediated by microglia, is crucial for the accumulation of intraneuronal amyloid and formation of neuritic plaques, suggesting that these two events are serially linked in a causal pathway leading to neurodegeneration and neuritic plaque formation. CSF1R inhibitors represent potential preventative or therapeutic approach that target the very earliest stages of the formation of intraneuronal amyloid and neuritic plaques.


Assuntos
Doença de Alzheimer/patologia , Aminopiridinas/farmacologia , Encéfalo/patologia , Microglia/efeitos dos fármacos , Neurônios/patologia , Pirróis/farmacologia , Doença de Alzheimer/metabolismo , Proteínas Amiloidogênicas/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores
13.
Circ Res ; 122(10): e75-e83, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29483093

RESUMO

RATIONALE: Disrupted proteostasis is one major pathological trait that heart failure (HF) shares with other organ proteinopathies, such as Alzheimer and Parkinson diseases. Yet, differently from the latter, whether and how cardiac preamyloid oligomers (PAOs) develop in acquired forms of HF is unclear. OBJECTIVE: We previously reported a rise in monophosphorylated, aggregate-prone desmin in canine and human HF. We now tested whether monophosphorylated desmin acts as the seed nucleating PAOs formation and determined whether positron emission tomography is able to detect myocardial PAOs in nongenetic HF. METHODS AND RESULTS: Here, we first show that toxic cardiac PAOs accumulate in the myocardium of mice subjected to transverse aortic constriction and that PAOs comigrate with the cytoskeletal protein desmin in this well-established model of acquired HF. We confirm this evidence in cardiac extracts from human ischemic and nonischemic HF. We also demonstrate that Ser31 phosphorylated desmin aggregates extensively in cultured cardiomyocytes. Lastly, we were able to detect the in vivo accumulation of cardiac PAOs using positron emission tomography for the first time in acquired HF. CONCLUSIONS: Ser31 phosphorylated desmin is a likely candidate seed for the nucleation process leading to cardiac PAOs deposition. Desmin post-translational processing and misfolding constitute a new, attractive avenue for the diagnosis and treatment of the cardiac accumulation of toxic PAOs that can now be measured by positron emission tomography in acquired HF.


Assuntos
Amiloide/metabolismo , Desmina/metabolismo , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , Processamento de Proteína Pós-Traducional , Amiloide/análise , Amiloide/efeitos dos fármacos , Animais , Catequina/análogos & derivados , Catequina/farmacologia , Células Cultivadas , Desmina/genética , Feminino , Vetores Genéticos , Insuficiência Cardíaca/etiologia , Humanos , Masculino , Espectrometria de Massas/métodos , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Isquemia Miocárdica/complicações , Fosforilação , Polimorfismo de Nucleotídeo Único , Tomografia por Emissão de Pósitrons/métodos , Pressão , Agregados Proteicos/efeitos dos fármacos , Dobramento de Proteína , Ratos , Proteínas Recombinantes/metabolismo , alfa-Cristalinas/deficiência , beta-Cristalinas/deficiência
14.
J Biol Chem ; 293(8): 2888-2902, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29282295

RESUMO

Amyloid-ß (Aß) and human islet amyloid polypeptide (hIAPP) aggregate to form amyloid fibrils that deposit in tissues and are associated with Alzheimer's disease (AD) and type II diabetes (T2D), respectively. Individuals with T2D have an increased risk of developing AD, and conversely, AD patients have an increased risk of developing T2D. Evidence suggests that this link between AD and T2D might originate from a structural similarity between aggregates of Aß and hIAPP. Using the cryoEM method microelectron diffraction, we determined the atomic structures of 11-residue segments from both Aß and hIAPP, termed Aß(24-34) WT and hIAPP(19-29) S20G, with 64% sequence similarity. We observed a high degree of structural similarity between their backbone atoms (0.96-Å root mean square deviation). Moreover, fibrils of these segments induced amyloid formation through self- and cross-seeding. Furthermore, inhibitors designed for one segment showed cross-efficacy for full-length Aß and hIAPP and reduced cytotoxicity of both proteins, although by apparently blocking different cytotoxic mechanisms. The similarity of the atomic structures of Aß(24-34) WT and hIAPP(19-29) S20G offers a molecular model for cross-seeding between Aß and hIAPP.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Modelos Moleculares , Emaranhados Neurofibrilares/metabolismo , Fragmentos de Peptídeos/metabolismo , Substituição de Aminoácidos , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular Tumoral , Biologia Computacional , Cristalografia por Raios X , Desenho de Fármacos , Células HEK293 , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/ultraestrutura , Polipeptídeo Amiloide das Ilhotas Pancreáticas/antagonistas & inibidores , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Camundongos , Microscopia Eletrônica de Transmissão , Mutação , Emaranhados Neurofibrilares/química , Emaranhados Neurofibrilares/efeitos dos fármacos , Emaranhados Neurofibrilares/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Nootrópicos/química , Nootrópicos/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Agregação Patológica de Proteínas/prevenção & controle , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura
15.
J Biol Chem ; 292(8): 3172-3185, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28049728

RESUMO

Most cases of Alzheimer's disease (AD) are sporadic, but a small percentage of AD cases, called familial AD (FAD), are associated with mutations in presenilin 1, presenilin 2, or the amyloid precursor protein. Amyloid precursor protein mutations falling within the amyloid-ß (Aß) sequence lead to a wide range of disease phenotypes. There is increasing evidence that distinct amyloid structures distinguished by amyloid conformation-dependent monoclonal antibodies have similarly distinct roles in pathology. It is possible that this phenotypic diversity of FAD associated with mutations within the Aß sequence is due to differences in the conformations adopted by mutant Aß peptides, but the effects of FAD mutations on aggregation kinetics and conformational and morphological changes of the Aß peptide are poorly defined. To gain more insight into this possibility, we therefore investigated the effects of 11 FAD mutations on the aggregation kinetics of Aß, as well as its ability to form distinct conformations recognized by a panel of amyloid conformation-specific monoclonal antibodies. We found that most FAD mutations increased the rate of aggregation of Aß. The FAD mutations also led to the adoption of alternative amyloid conformations distinguished by monoclonal antibodies and resulted in the formation of distinct aggregate morphologies as determined by transmission electron microscopy. In addition, several of the mutant peptides displayed a large reduction in thioflavin T fluorescence, despite forming abundant fibrils indicating that thioflavin T is a probe of conformational polymorphisms rather than a reliable indicator of fibrillization. Taken together, these results indicate that FAD mutations falling within the Aß sequence lead to dramatic changes in aggregation kinetics and influence the ability of Aß to form immunologically and morphologically distinct amyloid structures.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Mutação , Agregados Proteicos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/análise , Precursor de Proteína beta-Amiloide/ultraestrutura , Humanos , Conformação Proteica
16.
Elife ; 62017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28045370

RESUMO

hIAPP fibrils are associated with Type-II Diabetes, but the link of hIAPP structure to islet cell death remains elusive. Here we observe that hIAPP fibrils are cytotoxic to cultured pancreatic ß-cells, leading us to determine the structure and cytotoxicity of protein segments composing the amyloid spine of hIAPP. Using the cryoEM method MicroED, we discover that one segment, 19-29 S20G, forms pairs of ß-sheets mated by a dry interface that share structural features with and are similarly cytotoxic to full-length hIAPP fibrils. In contrast, a second segment, 15-25 WT, forms non-toxic labile ß-sheets. These segments possess different structures and cytotoxic effects, however, both can seed full-length hIAPP, and cause hIAPP to take on the cytotoxic and structural features of that segment. These results suggest that protein segment structures represent polymorphs of their parent protein and that segment 19-29 S20G may serve as a model for the toxic spine of hIAPP.


Assuntos
Amiloide/química , Amiloide/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Microscopia Crioeletrônica , Humanos , Conformação Proteica em Folha beta
17.
Biochem Biophys Res Commun ; 477(4): 700-705, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27363332

RESUMO

In Alzheimer's disease, soluble Aß oligomers are believed to play important roles in the disease pathogenesis, and their levels correlate with cognitive impairment. We have previously shown that Aß oligomers can be categorized into multiple structural classes based on their reactivity with conformation-dependent antibodies. In this study, we analyzed the structures of Aß40 oligomers belonging to two of these classes: fibrillar and prefibrillar oligomers. We found that fibrillar oligomers were similar in structure to fibrils but were less stable towards denaturation while prefibrillar oligomers were found to be partially disordered. These results are consistent with previously proposed structures for both oligomer classes while providing additional structural information.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/ultraestrutura , Amiloide/química , Amiloide/ultraestrutura , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/ultraestrutura , Dimerização , Conformação Proteica
18.
Proc Natl Acad Sci U S A ; 113(34): E4976-84, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27469165

RESUMO

Amyloid-ß (Aß) is present in humans as a 39- to 42-amino acid residue metabolic product of the amyloid precursor protein. Although the two predominant forms, Aß(1-40) and Aß(1-42), differ in only two residues, they display different biophysical, biological, and clinical behavior. Aß(1-42) is the more neurotoxic species, aggregates much faster, and dominates in senile plaque of Alzheimer's disease (AD) patients. Although small Aß oligomers are believed to be the neurotoxic species, Aß amyloid fibrils are, because of their presence in plaques, a pathological hallmark of AD and appear to play an important role in disease progression through cell-to-cell transmissibility. Here, we solved the 3D structure of a disease-relevant Aß(1-42) fibril polymorph, combining data from solid-state NMR spectroscopy and mass-per-length measurements from EM. The 3D structure is composed of two molecules per fibril layer, with residues 15-42 forming a double-horseshoe-like cross-ß-sheet entity with maximally buried hydrophobic side chains. Residues 1-14 are partially ordered and in a ß-strand conformation, but do not display unambiguous distance restraints to the remainder of the core structure.


Assuntos
Peptídeos beta-Amiloides/ultraestrutura , Fragmentos de Peptídeos/ultraestrutura , Peptídeos beta-Amiloides/genética , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Microscopia Eletrônica , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/genética , Conformação Proteica em Folha beta , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestrutura
19.
Neurol Neuroimmunol Neuroinflamm ; 3(3): e237, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27218118

RESUMO

BACKGROUND: Recent studies have implicated specific assembly subtypes of ß-amyloid (Aß) peptide, specifically soluble oligomers (soAß) as disease-relevant structures that may underlie memory loss in Alzheimer disease. Removing existing soluble and insoluble Aß assemblies is thought to be essential for any attempt at stabilizing brain function and slowing cognitive decline in Alzheimer disease. IV immunoglobulin (IVIg) therapies have been shown to contain naturally occurring polyclonal antibodies that recognize conformational neoepitopes of soluble or insoluble Aß assemblies including soAß. These naturally occurring polyclonal antibodies have been suggested to underlie the apparent clinical benefits of IVIg. However, direct evidence linking anti-Aß antibodies to the clinical bioactivity of IVIg has been lacking. METHODS: Five-month-old female Dutch APP E693Q mice were treated for 3 months with neat IVIg or with IVIg that had been affinity-depleted over immobilized Aß conformers in 1 of 2 assembly states. Memory was assessed in a battery of tests followed by quantification of brain soAß levels using standard anti-soAß antibodies. RESULTS: We provide evidence that NU4-type soAß (NU4-soAß) assemblies accumulate in the brains of Dutch APP E693Q mice and are associated with defects in memory, even in the absence of insoluble Aß plaques. Memory benefits were associated with depletion from APP E693Q mouse brain of NU4-soAß and A11-soAß but not OC-type fibrillar Aß oligomers. CONCLUSIONS: We propose that targeting of specific soAß assembly subtypes may be an important consideration in the therapeutic and/or prophylactic benefit of anti-Aß antibody drugs.

20.
Neurobiol Dis ; 82: 552-560, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26369878

RESUMO

Increased dietary consumption of docosahexaenoic acid (DHA) is associated with decreased risk for Alzheimer's disease (AD). These effects have been postulated to arise from DHA's pleiotropic effects on AD pathophysiology, including its effects on ß-amyloid (Aß) production, aggregation, and toxicity. While in vitro studies suggest that DHA may inhibit and reverse the formation of toxic Aß oligomers, it remains uncertain whether these mechanisms operate in vivo at the physiological concentrations of DHA attainable through dietary supplementation. We sought to clarify the effects of dietary DHA supplementation on Aß indices in a transgenic APP/PS1 rat model of AD. Animals maintained on a DHA-supplemented diet exhibited reductions in hippocampal Aß plaque density and modest improvements on behavioral testing relative to those maintained on a DHA-depleted diet. However, DHA supplementation also increased overall soluble Aß oligomer levels in the hippocampus. Further quantification of specific conformational populations of Aß oligomers indicated that DHA supplementation increased fibrillar (i.e. putatively less toxic) Aß oligomers and decreased prefibrillar (i.e. putatively more toxic) Aß oligomers. These results provide in vivo evidence suggesting that DHA can modulate Aß aggregation by stabilizing soluble fibrillar Aß oligomers and thus reduce the formation of both Aß plaques and prefibrillar Aß oligomers. However, since fibrillar Aß oligomers still retain inherent neurotoxicity, DHA may need to be combined with other interventions that can additionally reduce fibrillar Aß oligomer levels for more effective prevention of AD in clinical settings.


Assuntos
Doença de Alzheimer/dietoterapia , Peptídeos beta-Amiloides/metabolismo , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos , Hipocampo/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/dietoterapia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Placa Amiloide/psicologia , Presenilina-1/genética , Presenilina-1/metabolismo , Multimerização Proteica , Ratos Sprague-Dawley , Ratos Transgênicos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA