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.
Front Aging Neurosci ; 13: 640677, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33716717

RESUMO

Neurofibrillary tangles (NFTs) are a major pathologic hallmark of Alzheimer's disease (AD). Several studies have shown that amyloid ß oligomers (Aßo) and tau oligomers mediate their toxicity, in part, via binding to cellular prion protein (PrPC) and that some anti-PrP antibodies can block this interaction. We have generated a novel monoclonal anti-PrP antibody (TW1) and assessed the efficacy of passive immunization with it in a mouse model of AD with extensive tau pathology: hTau/PS1 transgenic (Tg) mice. These mice were injected intraperitoneally once a week with TW1 starting at 5 months of age. Behavior was assessed at 8 months of age and brain tissue was subsequently harvested for analysis of treatment efficacy at 9 months. Mice treated with TW1 did not show any significant difference in sensorimotor testing including traverse beam, rotarod, and locomotor activity compared to controls. Significant cognitive benefits were observed with the novel object recognition test (ORT) in the immunized mice (two-tailed, t-test p = 0.0019). Immunized mice also showed cognitive benefits on the closed field symmetrical maze (day 1 two-tailed t-test p = 0.0001; day 2 two-tailed t-test p = 0.0015; day 3 two-tailed t-test p = 0.0002). Reduction of tau pathology was observed with PHF-1 immunohistochemistry in the piriform cortex by 60% (two-tailed t-test p = 0.01) and in the dentate gyrus by 50% (two-tailed t-test p = 0.02) in animals treated with TW1 compared to controls. There were no significant differences in astrogliosis or microgliosis observed between treated and control mice. As assessed by Western blots using PHF-1, the TW1 therapy reduced phosphorylated tau pathology (two-tailed t-test p = 0.03) and improved the ratio of pathological soluble tau to tubulin (PHF1/tubulin; two-tailed t-test p = 0.0006). Reduction of tau pathology also was observed using the CP13 antibody (two-tailed t-test p = 0.0007). These results indicate that passive immunization with the TW1 antibody can significantly decrease tau pathology as assessed by immunohistochemical and biochemical methods, resulting in improved cognitive function in a tau transgenic mouse model of AD.

2.
Alzheimers Res Ther ; 10(1): 54, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29914551

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by physiologically endogenous proteins amyloid beta (Aß) and tau undergoing a conformational change and accumulating as soluble oligomers and insoluble aggregates. Tau and Aß soluble oligomers, which contain extensive ß-sheet secondary structure, are thought to be the most toxic forms. The objective of this study was to determine the ability of TWF9, an anti-ß-sheet conformation antibody (aßComAb), to selectively recognize pathological Aß and phosphorylated tau in AD human tissue compared with cognitively normal age-matched controls and to improve the performance of old 3xTg-AD mice with advanced pathology in behavioral testing after acute treatment with TWF9. METHODS: In this study, we used immunohistochemistry, immunoprecipitation, and enzyme-linked immunosorbent assay (ELISA) to characterize TWF9 specificity. We further assessed cognitive performance in old (18-22 months) 3xTg-AD mice using both a Barnes maze and novel object recognition after intraperitoneal administration of TWF9 (4 mg/kg) biweekly for 2 weeks before the start of behavioral testing. Injections continued for the duration of the behavioral testing, which lasted 2 weeks. RESULTS: Histological analysis of TWF9 in formalin-fixed paraffin-embedded human control and AD (ABC score: A3B3C3) brain tissue revealed preferential cytoplasmic immunoreactivity in neurons in the AD tissue compared with controls (p < 0.05). Furthermore, ELISA using oligomeric and monomeric Aß showed a preferential affinity for oligomeric Aß. Immunoprecipitation studies showed that TWF9 extracted both phosphorylated tau (p < 0.01) and Aß (p < 0.01) from fresh frozen brain tissues. Results show that treated old 3xTg-AD mice have an enhanced novel object recognition memory (p < 0.01) and Barnes maze performance (p = 0.05) compared with control animals. Overall plaque burden, neurofibrillary tangles, microgliosis, and astrocytosis remained unchanged. Soluble phosphorylated tau was significantly reduced in TWF9-treated mice (p < 0.05), and there was a trend for a reduction in soluble Aß levels in the brain homogenates of female 3xTg-AD mice (p = 0.06). CONCLUSIONS: This study shows that acute treatment with an aßComAb can effectively improve performance in behavioral testing without reduction of amyloid plaque burden, and that peripherally administered IgG can affect levels of pathological species in the brain.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/imunologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/terapia , Imunoterapia/métodos , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Proteínas de Ligação ao Cálcio , Cricetulus , Proteínas de Ligação a DNA/metabolismo , Feminino , Força da Mão , Humanos , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Mutação/genética , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
3.
J Alzheimers Dis ; 64(s1): S299-S312, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29562516

RESUMO

There is growing genetic and proteomic data highlighting the complexity of Alzheimer's disease (AD) pathogenesis. Greater use of unbiased "omics" approaches is being increasingly recognized as essential for the future development of effective AD research, that need to better reflect the multiple distinct pathway abnormalities that can drive AD pathology. The track record of success in AD clinical trials thus far has been very poor. In part, this high failure rate has been related to the premature translation of highly successful results in animal models that mirror only limited aspects of AD pathology to humans. We highlight our recent efforts to increase use of human tissue to gain a better understanding of the AD pathogenesis subtype variety and to develop several distinct therapeutic approaches tailored to address this diversity. These therapeutic approaches include the blocking of the Aß/apoE interaction, stimulation of innate immunity, and the simultaneous blocking of Aß/tau oligomer toxicity. We believe that future successful therapeutic approaches will need to be combined to better reflect the complexity of the abnormal pathways triggered in AD pathogenesis.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Animais , Humanos , Imunidade Inata , Proteoma
4.
Alzheimers Res Ther ; 10(1): 10, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29378642

RESUMO

BACKGROUND: Oligomeric forms of amyloid-ß (Aß) and tau are increasing being recognized as key toxins in the pathogenesis of Alzheimer's disease (AD). METHODS: We developed a novel monoclonal antibody (mAb), GW-23B7, that recognizes ß-sheet secondary structure on pathological oligomers of neurodegenerative diseases. RESULTS: The pentameric immunoglobulin M kappa chain (IgMκp) we developed specifically distinguishes intra- and extracellular pathology in human AD brains. Purified GW-23B7 showed a dissociation constant in the nanomolar range for oligomeric Aß and did not bind monomeric Aß. In enzyme-linked immunosorbent assays, it recognized oligomeric forms of both Aß and hyperphosphorylated tau. Aged triple-transgenic AD mice with both Aß and tau pathology infused intraperitoneally for 2 months showed IgMκp in the soluble brain homogenate, peaking at 24 h postinoculation. Treated mice exhibited significant cognitive rescue on radial arm maze testing compared with vehicle control-infused mice. Immunohistochemically, treatment resulted in a significant decrease of extracellular pathology. Biochemically, treatment resulted in significant reductions of oligomeric forms of Aß and tau. CONCLUSIONS: These results suggest that GW-23B7, an anti-ß-sheet conformational mAb humanized for clinical trials, may be an effective therapeutic agent for human AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Fatores Imunológicos/administração & dosagem , Conformação Proteica em Folha beta , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Imunoglobulina M , Aprendizagem em Labirinto , Camundongos Transgênicos , Atividade Motora , Proteínas tau/antagonistas & inibidores , Proteínas tau/química , Proteínas tau/imunologia
5.
Sci Rep ; 7(1): 8009, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808293

RESUMO

Inheritance of the apolipoprotein E4 (apoE4) genotype has been identified as the major genetic risk factor for late onset Alzheimer's disease (AD). Studies have shown that apoE, apoE4 in particular, binds to amyloid-ß (Aß) peptides at residues 12-28 of Aß and this binding modulates Aß accumulation and disease progression. We have previously shown in several AD transgenic mice lines that blocking the apoE/Aß interaction with Aß12-28 P reduced Aß and tau-related pathology, leading to cognitive improvements in treated AD mice. Recently, we have designed a small peptoid library derived from the Aß12-28 P sequence to screen for new apoE/Aß binding inhibitors with higher efficacy and safety. Peptoids are better drug candidates than peptides due to their inherently more favorable pharmacokinetic properties. One of the lead peptoid compounds, CPO_Aß17-21 P, diminished the apoE/Aß interaction and attenuated the apoE4 pro-fibrillogenic effects on Aß aggregation in vitro as well as apoE4 potentiation of Aß cytotoxicity. CPO_Aß17-21 P reduced Aß-related pathology coupled with cognitive improvements in an AD APP/PS1 transgenic mouse model. Our study suggests the non-toxic, non-fibrillogenic peptoid CPO_Aß17-21 P has significant promise as a new AD therapeutic agent which targets the Aß related apoE pathway, with improved efficacy and pharmacokinetic properties.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Peptoides/uso terapêutico , Animais , Linhagem Celular Tumoral , Cognição , Feminino , Humanos , Masculino , Camundongos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacocinética , Peptoides/química , Peptoides/farmacocinética , Ligação Proteica
6.
J Am Chem Soc ; 137(42): 13503-9, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26218347

RESUMO

Aggregation of amyloid ß-peptide (Aß) is implicated in the pathology of Alzheimer's disease (AD), with the soluble, Aß oligomeric species thought to be the critical pathological species. Identification and characterization of intermediate species formed during the aggregation process is crucial to the understanding of the mechanisms by which oligomeric species mediate neuronal toxicity and following disease progression. Probing these species proved to be extremely challenging, as evident by the lack of reliable sensors, due to their heterogeneous and transient nature. We describe here an oligomer-specific fluorescent chemical probe, BoDipy-Oligomer (BD-Oligo), developed through the use of the diversity-oriented fluorescent library approach (DOFLA) and high-content, imaging-based screening. This probe enables dynamic oligomer monitoring during fibrillogenesis in vitro and shows in vivo Aß oligomers staining possibility in the AD mice model.


Assuntos
Peptídeos beta-Amiloides/análise , Corantes Fluorescentes/química , Termodinâmica , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/classificação , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Camundongos , Modelos Moleculares
7.
PLoS One ; 6(9): e24844, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931860

RESUMO

Prion diseases currently have no effective therapy. These illnesses affect both animal and human populations, and are characterized by the conformational change of a normal self protein PrP(C) (C for cellular) to a pathological and infectious conformer, PrP(Sc) (Sc for scrapie). We used a well characterized tissue culture model of prion infection, where mouse neuroblastoma cells (N2a) were infected with 22L PrP(Sc), to screen compounds for anti-prion activity. In a prior study we designed a library of styryl based, potential imaging compounds which were selected for high affinity binding to Alzheimer's disease ß-amyloid plaques and good blood-brain barrier permeability. In the current study we screened this library for activity in the N2a/22L tissue culture system. We also tested the anti-prion activity of two clinically used drugs, trimipramine and fluphenazine, in the N2a/22L system. These were selected based on their structural similarity to quinacrine, which was previously reported to have anti-prion activity. All the compounds were also screened for toxicity in tissue culture and their ability to disaggregate amyloid fibrils composed of PrP and ß-amyloid synthetic peptides in vitro. Two of the imaging agents, 23I and 59, were found to be both effective at inhibiting prion infection in N2a/22L tissue culture and to be non-toxic. These two compounds, as well as trimipramine and fluphenazine were evaluated in vivo using wild-type CD-1 mice infected peripherally with 139A PrP(Sc). All four agents significantly prolonged the asymptomatic incubation period of prion infection (p<0.0001 log-rank test), as well as significantly reducing the degree of spongiform change, astrocytosis and PrP(Sc) levels in the brains of treated mice. These four compounds can be considered, with further development, as candidates for prion therapy.


Assuntos
Flufenazina/uso terapêutico , Doenças Priônicas/tratamento farmacológico , Trimipramina/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Camundongos , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Doenças Priônicas/metabolismo , Príons/metabolismo
8.
PLoS One ; 5(10): e13391, 2010 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-20967130

RESUMO

Many neurodegenerative diseases are characterized by the conformational change of normal self-proteins into amyloidogenic, pathological conformers, which share structural properties such as high ß-sheet content and resistance to degradation. The most common is Alzheimer's disease (AD) where the normal soluble amyloid ß (sAß) peptide is converted into highly toxic oligomeric Aß and fibrillar Aß that deposits as neuritic plaques and congophilic angiopathy. Currently, there is no highly effective treatment for AD, but immunotherapy is emerging as a potential disease modifying intervention. A major problem with most active and passive immunization approaches for AD is that both the normal sAß and pathogenic forms are equally targeted with the potential of autoimmune inflammation. In order to avoid this pitfall, we have developed a novel immunomodulatory method that specifically targets the pathological conformations, by immunizing with polymerized British amyloidosis (pABri) related peptide which has no sequence homology to Aß or other human proteins. We show that the pABri peptide through conformational mimicry induces a humoral immune response not only to the toxic Aß in APP/PS1 AD transgenic mice but also to paired helical filaments as shown on AD human tissue samples. Treated APP/PS1 mice had a cognitive benefit compared to controls (p<0.0001), associated with a reduction in the amyloid burden (p = 0.0001) and Aß40/42 levels, as well as reduced Aß oligomer levels. This type of immunomodulation has the potential to be a universal ß-sheet disrupter, which could be useful for the prevention or treatment of a wide range of neurodegenerative diseases.


Assuntos
Doença de Alzheimer/imunologia , Modelos Animais de Doenças , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos/sangue , Astrócitos/patologia , Western Blotting , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Locomoção , Aprendizagem em Labirinto , Camundongos , Microglia/patologia , Conformação Proteica , Teste de Desempenho do Rota-Rod
9.
Neurobiol Dis ; 34(2): 267-78, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19385058

RESUMO

The pathogenesis of prion diseases is related to conformational transformation of cellular prion protein (PrP(C)) into a toxic, infectious, and self-replicating conformer termed PrP(Sc). Following extracerebral inoculation, the replication of PrP(Sc) is confined for months to years to the lymporeticular system (LRS) before the secondary CNS involvement results in occurrence of neurological symptoms. Therefore, replication of PrP(Sc), in the early stage of infection can be targeted by therapeutic approaches, which like passive immunization have limited blood-brain-barrier penetration. In this study, we show that 6D11 anti-PrP monoclonal antibody (Mab) prevents infection on a FDC-P1 myeloid precursor cell line stably infected with 22L mouse adapted scrapie strain. Passive immunization of extracerebrally infected CD-1 mice with Mab 6D11 resulted in effective suppression of PrP(Sc) replication in the LRS. Although, a rebound of PrP(Sc) presence occurred when the Mab 6D11 treatment was stopped, passively immunized mice showed a prolongation of the incubation period by 36.9% (pb0.0001) and a significant decrease in CNS pathology compared to control groups receiving vehicle or murine IgG. Our results indicate that antibody-based therapeutic strategies can be used, even on a short-term basis, to delay or prevent disease in subjects accidentally exposed to prions.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunização Passiva/métodos , Sistema Linfático/efeitos dos fármacos , Células Progenitoras Mieloides/efeitos dos fármacos , Proteínas PrPSc/antagonistas & inibidores , Doenças Priônicas/tratamento farmacológico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/metabolismo , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Feminino , Humanos , Sistema Linfático/imunologia , Sistema Linfático/metabolismo , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/metabolismo , Proteínas PrPSc/biossíntese , Doenças Priônicas/imunologia , Doenças Priônicas/metabolismo , Conformação Proteica/efeitos dos fármacos , Scrapie/tratamento farmacológico , Scrapie/imunologia , Scrapie/metabolismo , Resultado do Tratamento
10.
J Virol ; 81(19): 10729-41, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652397

RESUMO

Prion diseases are fatal neurodegenerative disorders. Identification of possible therapeutic tools is important in the search for a potential treatment for these diseases. Congo red is an azo dye that has been used for many years to detect abnormal prion protein in the brains of diseased patients or animals. Congo red has little therapeutic potential for the treatment of these diseases due to toxicity and poor permeation of the blood-brain barrier. We have prepared two Congo red derivatives, designed without these liabilities, with potent activity in cellular models of prion disease. One of these compounds cured cells of the transmissible agent. The mechanism of action of these compounds is possibly multifactorial. The high affinity of Congo red derivatives, including compounds that are ineffective and are effective at the cure of prion disease, for abnormally folded prion protein suggests that the amyloidophylic property of these derivatives is not as critical to the mechanism of action as other effects. Congo red derivatives that are effective at the cure of prion disease increased the degradation of abnormal PrP by the proteasome. Therefore, the principal mechanism of action of the Congo red analogues was to prevent inhibition of proteasomal activity by PrPSc.


Assuntos
Derivados de Benzeno/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Proteínas PrPSc/antagonistas & inibidores , Doenças Priônicas/tratamento farmacológico , Scrapie/tratamento farmacológico , Animais , Derivados de Benzeno/química , Derivados de Benzeno/metabolismo , Compostos de Bifenilo/química , Compostos de Bifenilo/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Vermelho Congo/análogos & derivados , Camundongos , Camundongos Endogâmicos , Oxirredução , Proteínas PrPSc/análise , Proteínas PrPSc/metabolismo
11.
Arch Neurol ; 64(4): 595-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17420324

RESUMO

OBJECTIVE: To describe a novel molecular and pathological phenotype of Creutzfeldt-Jakob disease. Patient A 69-year-old woman with behavioral and personality changes followed by rapidly evolving dementia. RESULTS: Postmortem examination of the brain showed intracellular prion protein deposition and axonal swellings filled with amyloid fibrils. Biochemical analysis of the pathological prion protein disclosed a previously unrecognized PrP(Sc) tertiary structure lacking diglycosylated species. Genetic analysis revealed a wild-type prion protein gene. The prion agent responsible for this atypical phenotype was successfully passaged to bank voles. CONCLUSION: To our knowledge, our results define a new human prion disorder characterized by intracellular accumulation of a novel type of pathological prion protein.


Assuntos
Síndrome de Creutzfeldt-Jakob/metabolismo , Proteínas PrPSc/metabolismo , Idoso , Animais , Arvicolinae , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/transmissão , Evolução Fatal , Feminino , Genótipo , Glicosilação , Humanos , Immunoblotting , Espectrometria de Massas , Microscopia Imunoeletrônica , Fenótipo , Proteína PrP 27-30/química , Proteína PrP 27-30/genética , Proteína PrP 27-30/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/genética , Conformação Proteica , Estrutura Terciária de Proteína
12.
Proc Natl Acad Sci U S A ; 103(49): 18787-92, 2006 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-17116874

RESUMO

The amyloid-beta (Abeta) cascade hypothesis of Alzheimer's disease (AD) maintains that accumulation of Abeta peptide constitutes a critical event in the early disease pathogenesis. The direct binding between Abeta and apolipoprotein E (apoE) is an important factor implicated in both Abeta clearance and its deposition in the brain's parenchyma and the walls of meningoencephalic vessels as cerebral amyloid angiopathy. With the aim of testing the effect of blocking the apoE/Abeta interaction in vivo as a potential novel therapeutic target for AD pharmacotherapy, we have developed Abeta12-28P, which is a blood-brain-barrier-permeable nontoxic, and nonfibrillogenic synthetic peptide homologous to the apoE binding site on the full-length Abeta. Abeta12-28P binds with high affinity to apoE, preventing its binding to Abeta, but has no direct effect on Abeta aggregation. Abeta12-28P shows a strong pharmacological effect in vivo. Its systemic administration resulted in a significant reduction of Abeta plaques and cerebral amyloid angiopathy burden and a reduction of the total brain level of Abeta in two AD transgenic mice models. The treatment did not affect the levels of soluble Abeta fraction or Abeta oligomers, indicating that inhibition of the apoE/Abeta interaction in vivo has a net effect of increasing Abeta clearance over deposition and at the same time does not create conditions favoring formation of toxic oligomers. Furthermore, behavioral studies demonstrated that treatment with Abeta12-28P prevents a memory deficit in transgenic animals. These findings provide evidence of another therapeutic approach for AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Ligação Competitiva/fisiologia , Feminino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo
13.
J Alzheimers Dis ; 9(3 Suppl): 329-39, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16914871

RESUMO

Amyloid protein deposited in cerebral vessel walls and diffuse plaques of patients with hereditary cerebral hemorrhage with amyloidosis, Dutch type (HCHWA-D), is similar to the 40-42 residues amyloid beta (Abeta) in vessel walls and senile plaques in brains of patients with Alzheimer's disease (AD), Down's syndrome, and familial and sporadic cerebral amyloid angiopathy (CAA). In 1990 we sequenced the amyloid beta-protein precursor (AbetaPP) gene from HCHWA-D patients revealing a single mutation that results in an amino acid substitution, Abeta E22Q. Subsequent identification of additional mutations in the AbetaPP gene in familial AD (FAD) pedigrees revealed that whereas substitutions in the middle of Abeta, residues Abeta21-23, are predominantly vasculotropic, those found amino- or carboxyl-terminal to the Abeta sequence within AbetaPP enhance amyloid parenchymal plaque deposition. Studies of transfected cells showed that substitutions amino- or carboxyl-terminal to Abeta lead to either greater Abeta production or to enhanced secretion of the more hydrophobic thus more fibrillogenic Abeta1-42. Substitutions in the center of Abeta facilitate rapid aggregation and fibrillization, slower clearance across the blood-brain barrier and perivascular drainage to the systemic circulation, possibly higher resistance to proteolysis, and enhanced toxicity towards endothelial and smooth muscle cells. However, most AD patients have no genetic defects in AbetaPP, indicating that other factors may alter Abeta production, conformation, and/or clearance initiating the disease process.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Variação Genética/genética , Mutação Puntual/genética , Humanos , Países Baixos
14.
Eur J Neurosci ; 23(10): 2635-47, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16817866

RESUMO

Prion diseases are transmissible and invariably fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K (PK)-resistant conformer, scrapie PrP (PrP(Sc)). Humoral immunity may significantly prolong the incubation period and even prevent disease in murine models of prionoses. However, the mechanism(s) of action of anti-PrP monoclonal antibodies (Mabs) remain(s) obscure. The murine neuroblastoma N2a cell line, infected with the 22L mouse-adapted scrapie strain, was used to screen a large library of Mabs with similar binding affinities to PrP, to identify those antibodies which could clear established infection and/or prevent infection de novo. Three Mabs were found capable of complete and persistent clearing of already-infected N2a cells of PrP(Sc). These antibodies were 6D11 (generated to PK-resistant PrP(Sc) and detecting PrP residues 93-109), and 7H6 and 7A12, which were raised against recombinant PrP and react with neighbouring epitopes of PrP residues 130-140 and 143-155, respectively. Mabs were found to interact with PrP(Sc) formation both on the cell surface and after internalization in the cytosol. Treatment with Mabs was not associated with toxicity nor did it result in decreased expression of PrP(C). Both preincubation of N2a cells with Mabs prior to exposure to 22L inoculum and preincubation of the inoculum with Mabs prior to infecting N2a cells resulted in a significant reduction in PrP(Sc) levels. Information provided in these studies is important for the rational design of humoral immune therapy for prion infection in animals and eventually in humans.


Assuntos
Anticorpos Monoclonais/farmacologia , Proteínas PrPC/efeitos dos fármacos , Proteínas PrPSc/efeitos dos fármacos , Doenças Priônicas/imunologia , Doenças Priônicas/prevenção & controle , Animais , Afinidade de Anticorpos , Células Cultivadas , Mapeamento de Epitopos , Epitopos Imunodominantes , Camundongos , Proteínas PrPC/imunologia , Proteínas PrPSc/imunologia , Proteínas PrPSc/patogenicidade
15.
Methods Mol Biol ; 299: 221-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15980605

RESUMO

The characterization of proteins in their native state is essential for the understanding of patho-genic isoforms. A variant of the cysteine protease inhibitor cystatin C is the major constituent of the amyloid deposited in the cerebral vasculature of patients with the Icelandic form of hereditary cerebral hemorrhage with amyloidosis (HCHWA-I). In order to study the nature of the bio-physical changes owing to the Leu68Gln substitution in cystatin C, we have developed a purification procedure of human cystatin C in its native state. The protein is isolated from media of stably transfected tissue culture cells using physiological conditions that preclude protein denaturation. The importance of mild purification conditions is underscored by the finding that denaturation of the wild-type and variant proteins facilitates a similar folding of both molecules, diminishing their differences in structure and biophysical properties. Following native purification conditions, variant cystatin C has a distinct structure compared to the wild-type protein.


Assuntos
Cistatinas/isolamento & purificação , Western Blotting/métodos , Clonagem Molecular/métodos , Meios de Cultivo Condicionados/metabolismo , Cistatina C , Cistatinas/genética , Cistatinas/metabolismo , Humanos , Mutação , Análise de Sequência de Proteína/métodos , Transfecção/métodos
16.
J Biol Chem ; 279(37): 38936-42, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15247220

RESUMO

In prion diseases, the cellular prion protein (PrP(C)) is converted to an insoluble and protease-resistant abnormal isoform termed PrP(Sc). In different prion strains, PrP(Sc) shows distinct sites of endogenous or exogenous proteolysis generating a core fragment named PrP27-30. Sporadic Creutzfeldt-Jakob disease (sCJD), the most frequent human prion disease, clinically presents with a variety of neurological signs. As yet, the clinical variability observed in sCJD has not been fully explained by molecular studies relating two major types of PrP27-30 with unglycosylated peptides of 21 (type 1) and 19 kDa (type 2) and the amino acid methionine or valine at position 129. Recently, smaller C-terminal fragments migrating at 12 and 13 kDa have been detected in different sCJD phenotypes, but their significance remains unclear. By using two-dimensional immunoblot with anti-PrP antibodies, we identified two novel groups of protease-resistant PrP fragments in sCJD brain tissues. All sCJD cases with type 1 PrP27-30, in addition to MM subjects with type 2 PrP27-30, were characterized by the presence of unglycosylated PrP fragments of 16-17 kDa. Conversely, brain homogenates from patients VV and MV with type 2 PrP27-30 contained fully glycosylated PrP fragments, which after deglycosylation migrated at 17.5-18 kDa. Interestingly, PrP species of 17.5-18 kDa matched deglycosylated forms of the C1 PrP(C) fragment and were associated with tissue PrP deposition as plaque-like aggregates or amyloid plaques. These data show the presence of multiple PrP(Sc) conformations in sCJD and, in addition, shed new light on the correlation between sCJD phenotypes and disease-associated PrP molecules.


Assuntos
Síndrome de Creutzfeldt-Jakob/metabolismo , Príons/metabolismo , Adulto , Idoso , Sítios de Ligação , Western Blotting , Encéfalo/embriologia , Encéfalo/metabolismo , Detergentes/farmacologia , Eletroforese em Gel Bidimensional , Feminino , Glicosilação , Humanos , Immunoblotting , Imuno-Histoquímica , Focalização Isoelétrica , Masculino , Pessoa de Meia-Idade , Mutação , Peptídeos/química , Fenótipo , Proteínas PrPSc/química , Conformação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína
17.
J Biol Chem ; 278(47): 46199-202, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-14519758

RESUMO

The prion protein (PrP) binds copper and under some conditions copper can facilitate its folding into a more protease resistant form. Hence, copper levels may influence the infectivity of the scrapie form of prion protein (PrPSc). To determine the feasibility of copper-targeted therapy for prion disease, we treated mice with a copper chelator, D-(-)-penicillamine (D-PEN), starting immediately following intraperitoneal scrapie inoculation. D-PEN delayed the onset of prion disease in the mice by about 11 days (p = 0.002), and reduced copper levels in brain by 29% (p < 0.01) and in blood by 22% (p = 0.03) compared with control animals. Levels of other metals were not significantly altered in the blood or brain. Modest correlation was observed between incubation period and levels of copper in brain (p = 0.08) or blood (p = 0.04), indicating that copper levels are only one of many factors that influence the rate of progression of prion disease. In vitro, copper dose-dependently enhanced the proteinase K resistance of the prion protein, and this effect was counteracted in a dose-dependent manner by co-incubation with D-PEN. Overall, these findings indicate that copper levels can influence the conformational state of PrP, thereby enhancing its infectivity, and this effect can be attenuated by chelator-based therapy.


Assuntos
Quelantes/farmacologia , Quelantes/uso terapêutico , Cobre/metabolismo , Doenças Priônicas/prevenção & controle , Animais , Química Encefálica/efeitos dos fármacos , Terapia por Quelação/métodos , Cobre/análise , Cobre/sangue , Relação Dose-Resposta a Droga , Endopeptidase K/efeitos dos fármacos , Endopeptidase K/metabolismo , Camundongos , Camundongos Endogâmicos , Penicilamina/farmacologia , Penicilamina/uso terapêutico , Proteínas PrPSc/química , Proteínas PrPSc/efeitos dos fármacos , Proteínas PrPSc/metabolismo , Doenças Priônicas/tratamento farmacológico , Fatores de Tempo
18.
J Biol Chem ; 278(26): 23221-6, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12695513

RESUMO

Inherited amino acid substitutions at position 21, 22, or 23 of amyloid beta (Abeta) lead to presenile dementia or stroke. Insulin-degrading enzyme (IDE) can hydrolyze Abeta wild type, yet whether IDE is capable of degrading Abeta bearing pathogenic substitutions is not known. We studied the degradation of all of the published Abeta genetic variants by recombinant rat IDE (rIDE). Monomeric Abeta wild type, Flemish (A21G), Italian (E22K), and Iowa (D23N) variants were readily degraded by rIDE with a similar efficiency. However, proteolysis of Abeta Dutch (E22Q) and Arctic (E22G) was significantly lower as compared with Abeta wild type and the rest of the mutant peptides. In the case of Abeta Dutch, inefficient proteolysis was related to a high content of beta structure as assessed by circular dichroism. All of the Abeta variants were cleaved at Glu3-Phe4 and Phe4-Arg5 in addition to the previously described major sites within positions 13-15 and 18-21. SDS-stable Abeta dimers were highly resistant to proteolysis by rIDE regardless of the variant, suggesting that IDE recognizes a conformation that is available for interaction only in monomeric Abeta. These results raise the possibility that upregulation of IDE may promote the clearance of soluble Abeta in hereditary forms of Abeta diseases.


Assuntos
Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Insulisina/metabolismo , Animais , Hemorragia Cerebral/enzimologia , Hemorragia Cerebral/patologia , Demência/genética , Dimerização , Saúde da Família , Variação Genética , Humanos , Fragmentos de Peptídeos/análise , Ratos , Proteínas Recombinantes , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Acidente Vascular Cerebral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...