Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(2): e32737, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22393443

RESUMO

BACKGROUND: The identification of early mechanisms underlying Alzheimer's Disease (AD) and associated biomarkers could advance development of new therapies and improve monitoring and predicting of AD progression. Mitochondrial dysfunction has been suggested to underlie AD pathophysiology, however, no comprehensive study exists that evaluates the effect of different familial AD (FAD) mutations on mitochondrial function, dynamics, and brain energetics. METHODS AND FINDINGS: We characterized early mitochondrial dysfunction and metabolomic signatures of energetic stress in three commonly used transgenic mouse models of FAD. Assessment of mitochondrial motility, distribution, dynamics, morphology, and metabolomic profiling revealed the specific effect of each FAD mutation on the development of mitochondrial stress and dysfunction. Inhibition of mitochondrial trafficking was characteristic for embryonic neurons from mice expressing mutant human presenilin 1, PS1(M146L) and the double mutation of human amyloid precursor protein APP(Tg2576) and PS1(M146L) contributing to the increased susceptibility of neurons to excitotoxic cell death. Significant changes in mitochondrial morphology were detected in APP and APP/PS1 mice. All three FAD models demonstrated a loss of the integrity of synaptic mitochondria and energy production. Metabolomic profiling revealed mutation-specific changes in the levels of metabolites reflecting altered energy metabolism and mitochondrial dysfunction in brains of FAD mice. Metabolic biomarkers adequately reflected gender differences similar to that reported for AD patients and correlated well with the biomarkers currently used for diagnosis in humans. CONCLUSIONS: Mutation-specific alterations in mitochondrial dynamics, morphology and function in FAD mice occurred prior to the onset of memory and neurological phenotype and before the formation of amyloid deposits. Metabolomic signatures of mitochondrial stress and altered energy metabolism indicated alterations in nucleotide, Krebs cycle, energy transfer, carbohydrate, neurotransmitter, and amino acid metabolic pathways. Mitochondrial dysfunction, therefore, is an underlying event in AD progression, and FAD mouse models provide valuable tools to study early molecular mechanisms implicated in AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Mitocôndrias/metabolismo , Amiloide/genética , Animais , Biomarcadores/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Hipocampo/metabolismo , Humanos , Metabolômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Presenilina-1/genética , Fatores de Tempo
2.
J Neuropathol Exp Neurol ; 70(8): 653-61, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21760540

RESUMO

The relevance of cerebral amyloid angiopathy (CAA) to the pathogenesis of Alzheimer disease (AD) and dementia in general emphasizes the importance of developing novel targeting approaches for detecting and treating cerebrovascular amyloid (CVA) deposits. We developed a nanoparticle-based technology that uses a monoclonal antibody against fibrillar human amyloid-ß42 that is surface coated onto a functionalized phospholipid monolayer. We demonstrate that this conjugated nanoparticle binds to CVA deposits in arterioles of AD transgenic mice (Tg2576) after infusion into the external carotid artery using 3 different approaches. The first 2 approaches use a blood vessel enrichment of homogenized brain and a leptomeningeal vessel preparation from thin tangential brain slices from the surface of the cerebral cortex. Targeting of CVA by the antibody-coated nanoparticle was visualized using fluorescent lissamine rhodamine-labeled phospholipids in the nanoparticles, which were compared with fluorescent staining of the endothelial cells and amyloid deposits using confocal laser scanning microscopy. The third approach used high-field strength magnetic resonance imaging of antibody-coated iron oxide nanoparticles after infusion into the external carotid artery. Dark foci of contrast enhancement in cortical arterioles were observed in T2*-weighted images of ex vivo AD mouse brains that correlated histologically with CVA deposits. The targeting ability of these nanoparticles to CVA provides opportunities for the prevention and treatment of CAA.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Arteríolas/patologia , Compostos Férricos , Nanopartículas , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/patologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Arteríolas/ultraestrutura , Benzotiazóis , Modelos Animais de Doenças , Humanos , Imunoglobulina G , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Fragmentos de Peptídeos/imunologia , Placa Amiloide/ultraestrutura , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Tiazóis
3.
Curr Med Imaging Rev ; 7(1): 3-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21499442

RESUMO

A major objective in the treatment of Alzheimer's disease is amyloid plaque reduction. Transgenic mouse models of Alzheimer's disease provide a controlled and consistent environment for studying amyloid plaque deposition in Alzheimer's disease. Magnetic resonance imaging is an attractive tool for longitudinal studies because it offers non-invasive monitoring of amyloid plaques. Recent studies have demonstrated the ability of magnetic resonance imaging to detect individual plaques in living mice. This review discusses the mouse models, MR pulse sequences, and parameters that have been used to image plaques and how they can be optimized for future studies.

4.
PLoS One ; 6(12): e28881, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22216132

RESUMO

BACKGROUND: Therapeutic intervention of numerous brain-associated disorders currently remains unrealized due to serious limitations imposed by the blood-brain-barrier (BBB). The BBB generally allows transport of small molecules, typically <600 daltons with high octanol/water partition coefficients, but denies passage to most larger molecules. However, some receptors present on the BBB allow passage of cognate proteins to the brain. Utilizing such receptor-ligand systems, several investigators have developed methods for delivering proteins to the brain, a critical requirement of which involves covalent linking of the target protein to a carrier entity. Such covalent modifications involve extensive preparative and post-preparative chemistry that poses daunting limitations in the context of delivery to any organ. Here, we report creation of a 36-amino acid peptide transporter, which can transport a protein to the brain after routine intravenous injection of the transporter-protein mixture. No covalent linkage of the protein with the transporter is necessary. APPROACH: A peptide transporter comprising sixteen lysine residues and 20 amino acids corresponding to the LDLR-binding domain of apolipoprotein E (ApoE) was synthesized. Transport of beta-galactosidase, IgG, IgM, and antibodies against amyloid plques to the brain upon iv injection of the protein-transporter mixture was evaluated through staining for enzyme activity or micro single photon emission tomography (micro-SPECT) or immunostaining. Effect of the transporter on the integrity of the BBB was also investigated. PRINCIPAL FINDINGS: The transporter enabled delivery to the mouse brain of functional beta-galactosidase, human IgG and IgM, and two antibodies that labeled brain-associated amyloid beta plaques in a mouse model of Alzheimer's disease. SIGNIFICANCE: The results suggest the transporter is able to transport most or all proteins to the brain without the need for chemically linking the transporter to a protein. Thus, the approach offers an avenue for rapid clinical evaluation of numerous candidate drugs against neurological diseases including cancer. (299 words).


Assuntos
Amiloide/imunologia , Anticorpos/administração & dosagem , Imunoglobulina M/imunologia , beta-Galactosidase/administração & dosagem , Animais , Barreira Hematoencefálica , Camundongos , Tomografia Computadorizada de Emissão de Fóton Único
5.
PLoS One ; 5(1): e8813, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-20098681

RESUMO

A key question in understanding AD is whether extracellular Abeta deposition of parenchymal amyloid plaques or intraneuronal Abeta accumulation initiates the AD process. Amyloid precursor protein (APP) is endocytosed from the cell surface into endosomes where it is cleaved to produce soluble Abeta which is then released into the brain interstitial fluid. Intraneuronal Abeta accumulation is hypothesized to predominate from the neuronal uptake of this soluble extracellular Abeta rather than from ER/Golgi processing of APP. We demonstrate that substitution of the two adjacent histidine residues of Abeta40 results in a significant decrease in its binding with PC12 cells and mouse cortical/hippocampal neurons. These substitutions also result in a dramatic enhancement of both thioflavin-T positive fibril formation and binding to preformed Abeta fibrils while maintaining its plaque-binding ability in AD transgenic mice. Hence, alteration of the histidine domain of Abeta prevented neuronal binding and drove Abeta to enhanced fibril formation and subsequent amyloid plaque deposition--a potential mechanism for removing toxic species of Abeta. Substitution or even masking of these Abeta histidine residues might provide a new therapeutic direction for minimizing neuronal uptake and subsequent neuronal degeneration and maximizing targeting to amyloid plaques.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Animais , Córtex Cerebral/patologia , Hipocampo/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Células PC12 , Ratos , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície
6.
Biochemistry ; 48(43): 10405-15, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19775170

RESUMO

Several different monoclonal antibodies (mAbs) have been actively developed in the field of Alzheimer's disease (AD) for basic science and clinical applications; however, the binding kinetics of many of the mAbs with the beta-amyloid peptides (Abeta) are poorly understood. A panel of mAbs with different Abeta recognition sites, including our plaque-binding antibody (IgG4.1), a peptide-capturing antibody (11A50), and two classical mAbs (6E10 and 4G8) used for immunohistochemistry, were chosen for characterization of their kinetics of binding to monomeric and fibrillar forms of Abeta40 using surface plasmon resonance and their amyloid plaque binding ability in AD mouse brain sections using immunohistochemistry. The plaque-binding antibody (IgG4.1) with epitope specificity of Abeta(2-10) showed a weaker affinity (512 nM) for monomeric Abeta40 but a higher affinity (1.5 nM) for Abeta40 fibrils and labeled dense core plaques better than 6E10 as determined by immunohistochemistry. The peptide-capturing antibody (11A50) showed preferential affinity (32.5 nM) for monomeric Abeta40 but did not bind to Abeta40 fibrils, whereas antibodies 6E10 and 4G8 had moderate affinity for monomeric Abeta40 (22.3 and 30.1 nM, respectively). 4G8, which labels diffuse plaques better than 6E10, had a higher association rate constant than 6E10 but showed similar association and dissociation kinetics compared to those of 11A50. Enzymatic digestion of IgG4.1 to the F(ab')(2)4.1 fragments or their polyamine-modified derivatives that enhance blood-brain barrier permeability did not affect the kinetic properties of the antigen binding site. These differences in kinetic binding to monomeric and fibrillar Abeta among various antibodies could be utilized to distinguish mAbs that might be useful for immunotherapy or amyloid plaque imaging versus those that could be utilized for bioanalytical techniques.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Doença de Alzheimer/imunologia , Animais , Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Imuno-Histoquímica , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica
7.
Magn Reson Med ; 61(5): 1158-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19253386

RESUMO

One of the hallmark pathologies of Alzheimer's disease (AD) is amyloid plaque deposition. Plaques appear hypointense on T(2)-weighted and T(2)*-weighted MR images probably due to the presence of endogenous iron, but no quantitative comparison of various imaging techniques has been reported. We estimated the T(1), T(2), T(2)*, and proton density values of cortical plaques and normal cortical tissue and analyzed the plaque contrast generated by a collection of T(2)-weighted, T(2)*-weighted, and susceptibility-weighted imaging (SWI) methods in ex vivo transgenic mouse specimens. The proton density and T(1) values were similar for both cortical plaques and normal cortical tissue. The T(2) and T(2)* values were similar in cortical plaques, which indicates that the iron content of cortical plaques may not be as large as previously thought. Ex vivo plaque contrast was increased compared to a previously reported spin-echo sequence by summing multiple echoes and by performing SWI; however, gradient echo and SWI were found to be impractical for in vivo imaging due to susceptibility interface-related signal loss in the cortex.


Assuntos
Algoritmos , Doença de Alzheimer/diagnóstico , Imagem de Difusão por Ressonância Magnética/métodos , Modelos Animais de Doenças , Interpretação de Imagem Assistida por Computador/métodos , Placa Amiloide/patologia , Peptídeos beta-Amiloides/genética , Animais , Humanos , Aumento da Imagem/métodos , Camundongos , Camundongos Transgênicos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
8.
Neurochem Res ; 34(1): 102-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18373196

RESUMO

Accumulating evidence suggests that the conversion of Abeta peptides to soluble, neurotoxic polymers is the key event in the development of Alzheimer's disease (AD). Moreover, interactions between Abeta peptides and neuronal membrane lipids likely play a vital role in developing the neurotoxicity associated with AD. The aim of this study is to assess whether lipid matrix of neuronal membranes is affected by the accumulation of Abeta peptides in double transgenic mouse model of AD expressing both mutant human beta-amyloid precursor protein (APP) and presenilin 1 (PS1). We apply high pressure liquid chromatography with an evaporative light scattering detector to compare levels of cholesterol, galactocerebrosides, and phospholipid subclasses simultaneously in cortex samples between AD double transgenic mice at 4 months of age when Abeta production and amyloid plaque deposition is just beginning and at 9 months, when there is advanced Abeta levels and plaque deposition compared to age-matched wild-type (B6/SJL) mice. Both cholesterol (CL) and phospholipids (PL) are significantly lower in 9-month-old AD mice than the same age of B6/SJL mice. Among PL subclasses, phosphatidylethanolamine (PE), phosphatidylserine (PS) and phosphatidylcholine (PC) are selectively reduced in 9-month-old AD mice. The molar ratios of CL to PL in 9-month-old AD mice (1.19 +/- 0.27) were significantly higher than those of 9-month-old B6/SJL mice (0.81 +/- 0.08). In keeping with decreased levels of PL, there are also significant reductions of very long-chain n-3 fatty acids (docosahexaenoic acid) and n-6 fatty acid (arachidonic acid) in 9-month-old AD mice. On the other hand, ratios of total n-6 to total n-3 fatty acids were significantly higher in 9-month-old AD mice than in the same age of B6/SJL mice. Taken together, our present data support a role for the interactions of amyloid-beta peptide and neuronal membranes in the subsequent development of AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Córtex Cerebral/metabolismo , Lipídeos de Membrana/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Córtex Cerebral/patologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Camundongos , Camundongos Transgênicos , Presenilina-1/genética
9.
Pharm Res ; 25(8): 1861-72, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18443900

RESUMO

PURPOSE: The lack of an in vivo diagnostic test for AD has prompted the targeting of amyloid plaques with diagnostic imaging probes. We describe the development of a contrast agent (CA) for magnetic resonance microimaging that utilizes the F(ab')2 fragment of a monoclonal antibody raised against fibrillar human Abeta42 METHODS: This fragment is polyamine modified to enhance its BBB permeability and its ability to bind to amyloid plaques. It is also conjugated with a chelator and gadolinium for subsequent imaging of individual amyloid plaques RESULTS: Pharmacokinetic studies demonstrated this 125I-CA has higher BBB permeability and lower accumulation in the liver and kidney than F(ab')2 in WT mice. The CA retains its ability to bind Abeta40/42 monomers/fibrils and also binds to amyloid plaques in sections of AD mouse brain. Intravenous injection of 125I-CA into the AD mouse demonstrates targeting of amyloid plaques throughout the cortex/hippocampus as detected by emulsion autoradiography. Incubation of AD mouse brain slices in vitro with this CA resulted in selective enhancement on T1-weighted spin-echo images, which co-register with individual plaques observed on spatially matched T2-weighted spin-echo image CONCLUSIONS: Development of such a molecular probe is expected to open new avenues for the diagnosis of AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/imunologia , Meios de Contraste , Compostos Heterocíclicos , Compostos Organometálicos , Fragmentos de Peptídeos/imunologia , Placa Amiloide/patologia , Poliaminas , Animais , Cromatografia em Papel , Meios de Contraste/síntese química , Meios de Contraste/farmacocinética , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Compostos Heterocíclicos/síntese química , Compostos Heterocíclicos/farmacocinética , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos de Imunoglobulinas , Imunoglobulina G/imunologia , Camundongos , Camundongos Transgênicos , Compostos Organometálicos/síntese química , Compostos Organometálicos/farmacocinética
10.
Eur J Nucl Med Mol Imaging ; 35 Suppl 1: S82-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18239918

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is the most prevalent neurological condition affecting industrialized nations and will rapidly become a healthcare crisis as the population ages. Currently, the post-mortem histological observation of amyloid plaques and neurofibrillary tangles is the only definitive diagnosis available for AD. A pre-mortem biological or physiological marker specific for AD used in conjunction with current neurological and memory testing could add a great deal of confidence to the diagnosis of AD and potentially allow therapeutic intervention much earlier in the disease process. DISCUSSION AND CONCLUSION: Our group has developed MRI techniques to detect individual amyloid plaques in AD transgenic mouse brain in vivo. We are also developing contrast-enhancing agents to increase the specificity of detection of amyloid plaques. Such in vivo imaging of amyloid plaques will also allow the evaluation of anti-amyloid therapies being developed by the pharmaceutical industry in pre-clinical trials of AD transgenic mice. This short review briefly discusses our progress in these areas.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Aumento da Imagem/métodos , Imageamento por Ressonância Magnética/métodos , Placa Amiloide/metabolismo , Animais , Meios de Contraste , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Técnicas de Sonda Molecular
11.
J Neurochem ; 102(2): 420-33, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17596213

RESUMO

Targeting therapeutic or diagnostic proteins to the nervous system is limited by the presence of the blood-brain barrier. We report that a F(ab')(2) fragment of a monoclonal antibody against fibrillar human Abeta42 that is polyamine (p)-modified has increased permeability at the blood-brain barrier, comparable binding to the antigen, and comparable in vitro binding to amyloid plaques in Alzheimer's disease (AD) transgenic mouse brain sections. Intravenous injection of the pF(ab')(2)4.1 in the AD transgenic mouse demonstrated efficient targeting to amyloid plaques throughout the brain, whereas the unmodified fragment did not. Removal of the Fc portion of this antibody derivative will minimize the inflammatory response and cerebral hemorrhaging associated with passive immunization and provide increased therapeutic potential for treating AD. Coupling contrast agents/radioisotopes might facilitate the molecular imaging of amyloid plaques with magnetic resonance imaging/positron emission tomography. The efficient delivery of immunoglobulin G fragments may also have important applications to other neurodegenerative disorders or for the generalized targeting of nervous system antigens.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/metabolismo , Imunoterapia/métodos , Fragmentos de Peptídeos/imunologia , Placa Amiloide/imunologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Ligação Competitiva/imunologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Linhagem Celular Tumoral , Eletroforese em Gel de Poliacrilamida , Humanos , Fragmentos de Imunoglobulinas/uso terapêutico , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulina G/uso terapêutico , Imunoterapia/tendências , Injeções Intravenosas , Focalização Isoelétrica , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/antagonistas & inibidores , Peptídeo Hidrolases/química , Placa Amiloide/efeitos dos fármacos , Ligação Proteica/imunologia , Transporte Proteico/imunologia , Ensaio Radioligante , Receptores Imunológicos/efeitos dos fármacos , Receptores Imunológicos/metabolismo
12.
J Pharmacol Exp Ther ; 322(2): 541-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17505020

RESUMO

A novel magnetic resonance (MR) imaging contrast agent based on a derivative of human amyloid beta (Abeta) peptide, Gd[N-4ab/Q-4ab]Abeta 30, was previously shown to cross the blood-brain barrier (BBB) and bind to amyloid plaques in Alzheimer's disease (AD) transgenic mouse (APP/PS1) brain. We now report extensive plasma and brain pharmacokinetics of this contrast agent in wild-type (WT) and in APP/PS1 mice along with a quantitative summary of various physiological factors that govern its efficacy. Upon i.v. bolus administration, (125)I-Gd[N-4ab/Q-4ab]Abeta 30 was rapidly eliminated from the plasma following a three-exponential disposition, which is saturable at higher concentrations. Nevertheless, the contrast agent exhibited rapid and nonsaturable absorption at the BBB. The brain pharmacokinetic profile of (125)I-Gd[N-4ab/Q-4ab]Abeta 30 showed a rapid absorption phase followed by a slower elimination phase. No significant differences were observed in the plasma or brain kinetics of WT and APP/PS1 animals. Emulsion autoradiography studies conducted on WT and APP/PS1 mouse brain after an i.v. bolus administration of (125)I-Gd[N-4ab/Q-4ab]Abeta 30 in vivo confirmed the brain pharmacokinetic data and also demonstrated the preferential localization of the contrast agent on the plaques for an extended period of time. These attributes of the contrast agent are extremely useful in providing an excellent signal/noise ratio during longer MR scans, which may be essential for obtaining a high resolution image. In conclusion, this study documents the successful plaque targeting of Gd[N-4ab/Q-4ab]Abeta 30 and provides crucial pharmacokinetic information to determine the dose, mode of administration, and scan times for future in vivo MR imaging of amyloid plaques in AD transgenic mice.


Assuntos
Doença de Alzheimer/diagnóstico , Meios de Contraste/farmacocinética , Imageamento por Ressonância Magnética/métodos , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Meios de Contraste/química , Meios de Contraste/metabolismo , Gadolínio/química , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Cinética , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
13.
Neuroscientist ; 13(1): 38-48, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17229974

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia in the elderly. Cardinal pathologic features of AD are amyloid plaques and neurofibrillary tangles, and most in the field believe that the initiating events ultimately leading to clinical AD center on disordered metabolism of amyloid beta protein. Mouse models of AD have been created by inserting one or more human mutations associated with disordered amyloid metabolism and that cause early onset familial AD into the mouse genome. Human-like amyloid plaque formation increases dramatically with age in these transgenic mice. Amyloid reduction in humans is a major therapeutic objective, and AD transgenic mice allow controlled study of this biology. Recent work has shown that amyloid plaques as small as 35 microm can be detected using in vivo magnetic resonance microimaging (MRMI) at high magnetic field (9.4 T). In addition, age-dependent changes in metabolite concentration analogous to those that have been identified in human AD patients can be detected in these transgenic mice using single-voxel (1)H magnetic resonance spectroscopy ((1)H MRS) at high magnetic field. These MR-based techniques provide a new set of tools to the scientific community engaged in studying the biology of AD in transgenic models of the disease. For example, an obvious application is evaluating therapeutic modification of disease progression. Toward the end of this review, the authors include results from a pilot study demonstrating feasibility of using MRMI to detect therapeutic modification of plaque progression in AD transgenic mice.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Imageamento por Ressonância Magnética/métodos , Camundongos Transgênicos , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Humanos , Espectroscopia de Ressonância Magnética/métodos , Camundongos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Radioisótopos
14.
Neurobiol Aging ; 28(6): 921-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16781018

RESUMO

Aging is an intricate biological process thought to involve multiple molecular pathways. The spindle assembly checkpoint protein BubR1 has recently been implicated in aging since mutant mice that have small amounts of this protein (BubR1(H/H) mice) develop several early aging-associated phenotypes. The phenotype within the brain of BubR1(H/H) mice has not yet been established. Here we show that BubR1(H/H) mice exhibit features of age-related cerebral degeneration. We found that glial fibrillary acidic protein (GFAP), a marker of reactive astrogliosis, was expressed at increased levels in the cortex and thalamus of BubR1(H/H) mice as early as 1 month of age. Furthermore, CD11b, a marker of microgliosis, was markedly elevated in the cortex and hippocampus of BubR1(H/H) mice at 5 months of age. Levels of both GFAP and CD11b further increased with age. Our results demonstrate that BubR1 acts to prevent cerebral gliosis of both astrocytes and microglial cells, and suggest a role for BubR1 in the aging process of the brain.


Assuntos
Envelhecimento/fisiologia , Gliose/fisiopatologia , Proteínas Quinases/metabolismo , Fatores Etários , Análise de Variância , Animais , Antígeno CD11b/metabolismo , Proteínas de Ciclo Celular , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases
15.
J Pharmacol Exp Ther ; 318(1): 17-25, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16565169

RESUMO

Amyloid beta40 (Abeta40) and its derivatives are being developed as probes for the ante-mortem diagnosis of Alzheimer's disease. Putrescine-Abeta40 (PUT-Abeta40) showed better plaque targeting than the native Abeta40, which was not solely explained by the differences in their blood-brain-barrier (BBB) permeabilities. The objective of this study was to elucidate the physiological and biophysical factors influencing the differential targeting of Abeta40 and PUT-Abeta40. Despite better plaque-targeting ability 125I-PUT-Abeta40 was more rapidly cleared from the systemic circulation than amyloid beta40 labeled with 125I (125I-Abeta40) after i.v. administration in mice. The BBB permeability of both compounds was inhibited by circulating peripheral Abeta40 levels. 125I-Abeta40 but not 125I-PUT-Abeta40 was actively taken up by the mouse brain slices in vitro. Only fluorescein-Abeta40, not fluorescein-PUT-Abeta40, was localized in the brain parenchymal cells in vitro. The metabolism of 125I-Abeta40 in the brain slices was twice as great as 125I-PUT-Abeta40. 125I-Abeta40 efflux from the brain slices was saturable and found to be 5 times greater than that of 125I-PUT-Abeta40. Thioflavin-T fibrillogenesis assay demonstrated that PUT-Abeta40 has a greater propensity to form insoluble fibrils compared with Abeta40, most likely due to the ability of PUT-Abeta40 to form beta sheet structure more readily than Abeta40. These results demonstrate that the inadequate plaque targeting of Abeta40 is due to cellular uptake, metabolism, and efflux from the brain parenchyma. Despite better plaque targeting of PUTAbeta40, its propensity to form fibrils may render it less suitable for human use and thus allow increased focus on the development of novel derivatives of Abeta with improved characteristics.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/fisiologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/fisiologia , Putrescina/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Humanos , Camundongos , Fragmentos de Peptídeos/farmacologia , Placa Amiloide/efeitos dos fármacos , Putrescina/farmacologia
16.
J Neurosci ; 25(43): 10041-8, 2005 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-16251453

RESUMO

The ability to detect individual Alzheimer's amyloid plaques in vivo by magnetic resonance microimaging (MRI) should improve diagnosis and also accelerate discovery of effective therapeutic agents for Alzheimer's disease (AD). Here, we perform in vivo and ex vivo MRI on double transgenic AD mice as well as wild-type mice at varying ages and correlate these with thioflavin-S and iron staining histology. Quantitative counts of individual plaques on MRI increase with age and correlate with histologically determined plaque burden. Plaques 20 microm in diameter can be detected in AD mice as young as 3 months of age with ex vivo MRI. Plaques 35 microm in diameter can be detected by 9 months of age with in vivo MRI. In vivo MRI of individual Alzheimer's amyloid plaques provides a noninvasive estimate of plaque burden in transgenic AD mice that might be useful in assessing the efficacy of amyloid reduction therapies.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Imageamento por Ressonância Magnética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Fatores Etários , Animais , Benzotiazóis , Mapeamento Encefálico , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Coloração e Rotulagem/métodos , Tiazóis/metabolismo
17.
Proc Natl Acad Sci U S A ; 102(33): 11906-10, 2005 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-16091461

RESUMO

Currently no definitive biomarker of Alzheimer's disease (AD) is available, and this impedes both clinical diagnosis in humans and drug discovery in transgenic animal models. Proton magnetic resonance spectroscopy ((1)H MRS) provides a noninvasive way to investigate in vivo neurochemical abnormalities. Each observable metabolite can potentially provide information about unique in vivo pathological processes at the molecular or cellular level. In this study, the age-dependent 1H MRS profile of transgenic AD mice was compared to that of wild-type mice. Twenty-seven APP-PS1 mice (which coexpress mutated human presenilin 1 and amyloid-beta precursor protein) and 30 wild-type mice age 66-904 days were examined, some repeatedly. A reduction in the levels of N-acetylaspartate and glutamate, compared with total creatine levels, was found in APP-PS1 mice with advancing age. The most striking finding was a dramatic increase in the concentration of myo-inositol with age in APP-PS1 mice, which was not observed in wild-type mice. The age-dependent neurochemical changes observed in APP-PS1 mice agree with results obtained from in vivo human MRS studies. Among the different transgenic mouse models of AD that have been studied with 1H MRS, APP-PS1 mice seem to best match the neurochemical profile exhibited in human AD. 1H MRS could serve as a sensitive in vivo surrogate indicator of therapeutic efficacy in trials of agents designed to reduce neurotoxicity due to microglial activation. Because of its noninvasive and repeatable nature, MRS in transgenic models of AD could substantially accelerate drug discovery for this disease.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Modelos Animais de Doenças , Animais , Progressão da Doença , Feminino , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Prótons
18.
Brain Res ; 1045(1-2): 185-98, 2005 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-15910777

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder, characterized by the degeneration of upper and lower motor neurons (MNs). Central nervous system features include a loss of Betz cells and other pyramidal cells from sensorimotor cortex. The intrinsic mechanism underlying this selective motor neuron loss has not been identified. A recent in vitro study has provided evidence of a novel programmed cell death (PCD) pathway that is unique to spinal cord MNs and is exacerbated by superoxide dismutase (SOD) mutations. This PCD pathway is triggered through the Fas receptor and involves the apoptosis signal-regulating kinase 1 (ASK1), the p38 MAP kinase, and the neuronal form of nitric oxide synthase (nNOS). Previously, we found significant increases in the numbers of ventral horn MNs immunopositive for these enzymes in the spinal cords of mutant SOD transgenic (G93A) mice as early as 60 days of age, suggesting that this pathway may be active in vivo. Since the upper MNs of ALS patients and G93A mice are also known to degenerate, the purpose of the present study was to investigate the possible activation of this PCD pathway in the MNs of the sensorimotor cortex of G93A transgenic mice. Compared to non-transgenic littermates, the G93A mice showed significant increases in the numbers of MNs immunopositive for the active (phosphorylated) forms of ASK1, p38, MKK3/6 (the known activator of p38), and also active caspase-3, as early as 60 days of age. Another stress-activated protein kinase, c-Jun N-terminal kinase (JNK), commonly activated in other neurodegenerative disorders such as Alzheimer's disease, showed no increases in G93A mice at any age. These results suggest that, not only has a PCD pathway been activated in the cortical MNs, but one that may be unique to ALS. Moreover, these findings suggest that earlier diagnosis and therapeutic intervention may be possible for successful treatment of ALS. Consequently, these enzymes may provide the biochemical markers to enable earlier diagnosis of ALS and molecular targets for the development of new therapeutic compounds.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Apoptose/fisiologia , Córtex Motor/enzimologia , Neurônios Motores/enzimologia , Estresse Oxidativo/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Caspase 3 , Caspases/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática/fisiologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 3 , MAP Quinase Quinase Quinase 5/metabolismo , Camundongos , Camundongos Transgênicos , Córtex Motor/patologia , Córtex Motor/fisiopatologia , Neurônios Motores/patologia , Mutação/genética , Fosforilação , Transdução de Sinais/fisiologia , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Regulação para Cima/fisiologia
19.
J Pharmacol Exp Ther ; 313(3): 1370-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15743932

RESUMO

Amyloid plaques are formed in the extracellular space of Alzheimer's disease (AD) brain due to the accumulation of amyloid beta (Abeta) proteins such as Abeta40. The relationship between Abeta40 pharmacokinetics and its accumulation within and clearance from the brain in both wild-type (WT) and AD transgenic mice (APP,PS1) was studied to understand the mechanism of amyloid plaque formation and the potential use of Abeta40 as a probe to target and detect amyloid plaques. In both WT and APP,PS1 mice, the (125)I-Abeta40 tracer exhibited biexponential disposition in plasma with very short first and second phase half-lives. The (125)I-Abeta40 was significantly metabolized in the liver kidney > spleen. Coadministration of exogenous Abeta40 inhibited the plasma clearance and the uptake of (125)I-Abeta40 at the blood-brain barrier (BBB) in WT animals but did not affect its elimination from the brain. The (125)I-Abeta40 was shown to be metabolized within and effluxed from the brain parenchyma. The rate of efflux from APP,PS1 brain slices was substantially lower compared with WT brain slices. Since the Abeta40 receptor at the BBB can be easily saturated, the blood-to-brain transport of Abeta40 is less likely to be a primary contributor to the amyloid plaque formation in APP,PS1 mice. The decreased elimination of Abeta40 from the brain is most likely responsible for the amyloid plaque formation in the brain of APP,PS1 mice. Furthermore, inadequate targeting of Abeta40 to amyloid plaques, despite its high BBB permeability, is due to the saturability of Abeta40 transporter at the BBB and its metabolism and efflux from the brain.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacocinética , Barreira Hematoencefálica , Fragmentos de Peptídeos/farmacocinética , Placa Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Transporte Biológico , Encéfalo/metabolismo , Humanos , Radioisótopos do Iodo , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1
20.
Magn Reson Med ; 52(6): 1263-71, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15562496

RESUMO

One of the cardinal pathologic features of Alzheimer's disease (AD) is the formation of senile, or amyloid, plaques. Transgenic mice have been developed that express one or more of the genes responsible for familial AD in humans. Doubly transgenic mice develop "human-like" plaques, providing a mechanism to study amyloid plaque biology in a controlled manner. Imaging of labeled plaques has been accomplished with other modalities, but only MRI has sufficient spatial and contrast resolution to visualize individual plaques noninvasively. Methods to optimize visualization of plaques in vivo in transgenic mice at 9.4 T using a spin echo sequence based on adiabatic pulses are described. Preliminary results indicate that a spin echo acquisition more accurately reflects plaque size, while a T2* weighted gradient echo sequence reflects plaque iron content, not plaque size. In vivo MRI-ex vivo MRI-in vitro histologic correlations are provided. Histologically verified plaques as small as 50 microm in diameter were visualized in living animals. To our knowledge this work represents the first demonstration of noninvasive in vivo visualization of individual AD plaques without the use of a contrast agent.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Placa Amiloide/patologia , Animais , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA