Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Pharm ; 21(5): 2176-2186, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38625027

RESUMO

The blood-brain barrier (BBB) is instrumental in clearing toxic metabolites from the brain, such as amyloid-ß (Aß) peptides, and in delivering essential nutrients to the brain, like insulin. In Alzheimer's disease (AD) brain, increased Aß levels are paralleled by decreased insulin levels, which are accompanied by insulin signaling deficits at the BBB. Thus, we investigated the impact of insulin-like growth factor and insulin receptor (IGF1R and IR) signaling on Aß and insulin trafficking at the BBB. Following intravenous infusion of an IGF1R/IR kinase inhibitor (AG1024) in wild-type mice, the BBB trafficking of 125I radiolabeled Aß peptides and insulin was assessed by dynamic SPECT/CT imaging. The brain efflux of [125I]iodo-Aß42 decreased upon AG1024 treatment. Additionally, the brain influx of [125I]iodoinsulin, [125I]iodo-Aß42, [125I]iodo-Aß40, and [125I]iodo-BSA (BBB integrity marker) was decreased, increased, unchanged, and unchanged, respectively, upon AG1024 treatment. Subsequent mechanistic studies were performed using an in vitro BBB cell model. The cell uptake of [125I]iodoinsulin, [125I]iodo-Aß42, and [125I]iodo-Aß40 was decreased, increased, and unchanged, respectively, upon AG1024 treatment. Further, AG1024 reduced the phosphorylation of insulin signaling kinases (Akt and Erk) and the membrane expression of Aß and insulin trafficking receptors (LRP-1 and IR-ß). These findings reveal that insulin signaling differentially regulates the BBB trafficking of Aß peptides and insulin. Moreover, deficits in IGF1R and IR signaling, as observed in the brains of type II diabetes and AD patients, are expected to increase Aß accumulation while decreasing insulin delivery to the brain, which has been linked to the progression of cognitive decline in AD.


Assuntos
Peptídeos beta-Amiloides , Barreira Hematoencefálica , Insulina , Transdução de Sinais , Animais , Masculino , Camundongos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Insulina/metabolismo , Radioisótopos do Iodo , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tirfostinas/farmacologia
2.
J Pharmacol Exp Ther ; 386(1): 102-110, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37221092

RESUMO

Plasma pharmacokinetic (PK) data are required as an input function for graphical analysis of single positron emission computed tomography/computed tomography (SPECT/CT) and positron emission tomography/CT (PET/CT) data to evaluate tissue influx rate of radiotracers. Dynamic heart imaging data are often used as a surrogate of plasma PK. However, accumulation of radiolabel in the heart tissue may cause overprediction of plasma PK. Therefore, we developed a compartmental model, which involves forcing functions to describe intact and degraded radiolabeled proteins in plasma and their accumulation in heart tissue, to deconvolve plasma PK of 125I-amyloid beta 40 (125I-Aß 40) and 125I-insulin from their dynamic heart imaging data. The three-compartment model was shown to adequately describe the plasma concentration-time profile of intact/degraded proteins and the heart radioactivity time data obtained from SPECT/CT imaging for both tracers. The model was successfully applied to deconvolve the plasma PK of both tracers from their naïve datasets of dynamic heart imaging. In agreement with our previous observations made by conventional serial plasma sampling, the deconvolved plasma PK of 125I-Aß 40 and 125I-insulin in young mice exhibited lower area under the curve than aged mice. Further, Patlak plot parameters extracted using deconvolved plasma PK as input function successfully recapitulated age-dependent plasma-to-brain influx kinetics changes. Therefore, the compartment model developed in this study provides a novel approach to deconvolve plasma PK of radiotracers from their noninvasive dynamic heart imaging. This method facilitates the application of preclinical SPECT/PET imaging data to characterize distribution kinetics of tracers where simultaneous plasma sampling is not feasible. SIGNIFICANCE STATEMENT: Knowledge of plasma pharmacokinetics (PK) of a radiotracer is necessary to accurately estimate its plasma-to-brain influx. However, simultaneous plasma sampling during dynamic imaging procedures is not always feasible. In the current study, we developed approaches to deconvolve plasma PK from dynamic heart imaging data of two model radiotracers, 125I-amyloid beta 40 (125I-Aß 40) and 125I-insulin. This novel method is expected to minimize the need for conducting additional plasma PK studies and allow for accurate estimation of the brain influx rate.


Assuntos
Insulinas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Animais , Camundongos , Peptídeos beta-Amiloides , Elétrons , Tomografia Computadorizada por Raios X , Tomografia por Emissão de Pósitrons/métodos
3.
J Alzheimers Dis ; 85(3): 1031-1044, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34924382

RESUMO

BACKGROUND: Age is the most common risk factor for Alzheimer's disease (AD), a neurodegenerative disorder characterized by the hallmarks of toxic amyloid-ß (Aß) plaques and hyperphosphorylated tau tangles. Moreover, sub-physiological brain insulin levels have emerged as a pathological manifestation of AD. OBJECTIVE: Identify age-related changes in the plasma disposition and blood-brain barrier (BBB) trafficking of Aß peptides and insulin in mice. METHODS: Upon systemic injection of 125I-Aß40, 125I-Aß42, or 125I-insulin, the plasma pharmacokinetics and brain influx were assessed in wild-type (WT) or AD transgenic (APP/PS1) mice at various ages. Additionally, publicly available single-cell RNA-Seq data [GSE129788] was employed to investigate pathways regulating BBB transport in WT mice at different ages. RESULTS: The brain influx of 125I-Aß40, estimated as the permeability-surface area product, decreased with age, accompanied by an increase in plasma AUC. In contrast, the brain influx of 125I-Aß42 increased with age, accompanied by a decrease in plasma AUC. The age-dependent changes observed in WT mice were accelerated in APP/PS1 mice. As seen with 125I-Aß40, the brain influx of 125I-insulin decreased with age in WT mice, accompanied by an increase in plasma AUC. This finding was further supported by dynamic single-photon emission computed tomography (SPECT/CT) imaging studies. RAGE and PI3K/AKT signaling pathways at the BBB, which are implicated in Aß and insulin transcytosis, respectively, were upregulated with age in WT mice, indicating BBB insulin resistance. CONCLUSION: Aging differentially affects the plasma pharmacokinetics and brain influx of Aß isoforms and insulin in a manner that could potentially augment AD risk.


Assuntos
Envelhecimento , Doença de Alzheimer , Peptídeos beta-Amiloides/farmacocinética , Barreira Hematoencefálica/metabolismo , Insulina/farmacocinética , Placa Amiloide/metabolismo , Fatores Etários , Envelhecimento/sangue , Envelhecimento/fisiologia , Doença de Alzheimer/sangue , Doença de Alzheimer/patologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Modelos Animais de Doenças , Radioisótopos do Iodo/farmacocinética , Camundongos , Camundongos Transgênicos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único
4.
Mol Pharm ; 18(11): 4148-4161, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34664956

RESUMO

Elevated exposure to toxic amyloid beta (Aß) peptides and consequent blood-brain barrier (BBB) dysfunction are believed to promote vasculopathy in Alzheimer's disease (AD). However, the accumulation kinetics of different Aß isoforms within the BBB endothelium and how it drives BBB dysfunction are not clearly characterized. Using single positron emission computed tomography (SPECT)-computed tomography (CT) dynamic imaging coupled with population pharmacokinetic modeling, we investigated the accumulation kinetics of Aß40 and Aß42 in the BBB endothelium. Brain clearance was quantified after intracerebral administration of 125I-Aß, and BBB-mediated transport was shown to account for 54% of 125I-Aß40 total clearance. A brain influx study demonstrated lower values of both maximal rate (Vmax) and Michaelis constant (Km) for 125I-Aß42 compared to 125I-Aß40. Validated by a transcytosis study in polarized human BBB endothelial cell (hCMEC/D3) monolayers, model simulations demonstrated impaired exocytosis was responsible for inefficient permeability and enhanced accumulation of Aß42 in the BBB endothelium. Further, both isoforms were shown to disrupt the exocytosis machinery of BBB endothelial cells so that a vicious cycle could be generated. The validated model was able to capture changes in Aß steady-state levels in plasma as well as the brain during AD progression and allowed us to predict the kinetics of Aß accumulation in the BBB endothelium.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Doença de Alzheimer/diagnóstico , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/diagnóstico por imagem , Linhagem Celular , Modelos Animais de Doenças , Humanos , Camundongos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Transcitose
5.
J Pharmacol Exp Ther ; 376(3): 482-490, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33303699

RESUMO

Blood-brain barrier (BBB) endothelial cells lining the cerebral microvasculature maintain dynamic equilibrium between soluble amyloid-ß (Aß) levels in the brain and plasma. The BBB dysfunction prevalent in Alzheimer disease contributes to the dysregulation of plasma and brain Aß and leads to the perturbation of the ratio between Aß42 and Aß40, the two most prevalent Aß isoforms in patients with Alzheimer disease. We hypothesize that BBB endothelium distinguishes between Aß40 and Aß42, distinctly modulates their trafficking kinetics between plasma and brain, and thereby contributes to the maintenance of healthy Aß42/Aß40 ratios. To test this hypothesis, we investigated Aß40 and Aß42 trafficking kinetics in hCMEC/D3 monolayers (human BBB cell culture model) in vitro as well as in mice in vivo. Although the rates of uptake of fluorescein-labeled Aß40 and Aß42 (F-Aß40 and F-Aß42) were not significantly different on the abluminal side, the luminal uptake rate of F-Aß42 was substantially higher than F-Aß40. Since higher plasma Aß levels were shown to aggravate BBB dysfunction and trigger cerebrovascular disease, we systematically investigated the dynamic interactions of luminal [125I]Aß peptides and their trafficking kinetics at BBB using single-photon emission computed tomography/computed tomography imaging in mice. Quantitative modeling of the dynamic imaging data thus obtained showed that the rate of uptake of toxic [125I]Aß42 and its subsequent BBB transcytosis is significantly higher than [125I]Aß40. It is likely that the molecular mechanisms underlying these kinetic differences are differentially affected in Alzheimer and cerebrovascular diseases, impact plasma and brain levels of Aß40 and Aß42, engender shifts in the Aß42/Aß40 ratio, and unleash downstream toxic effects. SIGNIFICANCE STATEMENT: Dissecting the binding and uptake kinetics of Aß40 and Aß42 at the BBB endothelium will facilitate the estimation of Aß40 versus Aß42 exposure to the BBB endothelium and allow assessment of the risk of BBB dysfunction by monitoring Aß42 and Aß40 levels in plasma. This knowledge, in turn, will aid in elucidating the role of these predominant Aß isoforms in aggravating BBB dysfunction and cerebrovascular disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Fragmentos de Peptídeos/metabolismo , Linhagem Celular , Endotélio/metabolismo , Humanos , Cinética , Modelos Biológicos , Transporte Proteico
6.
J Pharmacol Exp Ther ; 375(2): 308-316, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32778535

RESUMO

Treatments to elevate high-density lipoprotein (HDL) levels in plasma have decreased cerebrovascular amyloid -ß (Aß) deposition and mitigated cognitive decline in Alzheimer disease (AD) transgenic mice. Since the major protein component of HDL particles, apolipoprotein A-I (ApoA-I), has very low permeability at the blood-brain barrier (BBB), we investigated 4F, an 18-amino-acid ApoA-I/HDL mimetic peptide, as a therapeutic alternative. Specifically, we examined the BBB permeability of 4F and its effects on [125I]Aß trafficking from brain to blood and from blood to brain. After systemic injection in mice, the BBB permeability of [125I]4F, estimated as the permeability-surface area (PS) product, ranged between 2 and 5 × 10-6 ml/g per second in various brain regions. The PS products of [125I]4F were ∼1000-fold higher compared with those determined for [125I]ApoA-I. Moreover, systemic infusion with 4F increased the brain efflux of intracerebrally injected [125I]Aß42. Conversely, 4F infusion decreased the brain influx of systemically injected [125I]Aß42. Interestingly, 4F did not significantly alter the brain influx of [125I]Aß40. To corroborate the in vivo findings, we evaluated the effects of 4F on [125I]Aß42 transcytosis across polarized human BBB endothelial cell (hCMEC/D3) monolayers. Treatment with 4F increased the abluminal-to-luminal flux and decreased the luminal-to-abluminal flux of [125I]Aß42 across the hCMEC/D3 monolayers. Additionally, 4F decreased the endothelial accumulation of fluorescein-labeled Aß42 in the hCMEC/D3 monolayers. These findings provide a mechanistic interpretation for the reductions in brain Aß burden reported in AD mice after oral 4F administration, which represents a novel strategy for treating AD and cerebral amyloid angiopathy. SIGNIFICANCE STATEMENT: The brain permeability of the ApoA-I mimetic peptide 4F was estimated to be ∼1000-fold greater than ApoA-I after systemic injection of radiolabeled peptide/protein in mice. Further, 4F treatment increased the brain efflux of amyloid -ß and also decreased its brain influx, as evaluated in mice and in blood-brain barrier cell monolayers. Thus, 4F represents a potential therapeutic strategy to mitigate brain amyloid accumulation in cerebral amyloid angiopathy and Alzheimer disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Peptídeos/farmacologia , Peptídeos beta-Amiloides/sangue , Animais , Camundongos , Fragmentos de Peptídeos/sangue , Transporte Proteico/efeitos dos fármacos
7.
J Pharmacol Exp Ther ; 369(3): 481-488, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30971477

RESUMO

Recent studies suggest that apolipoprotein A-I (ApoA-I), the major protein constituent of high-density lipoprotein particles, plays a critical role in preserving cerebrovascular integrity and reducing Alzheimer's risk. ApoA-I present in brain is thought to be primarily derived from the peripheral circulation. Although plasma-to-brain delivery of ApoA-I is claimed to be handled by the blood-cerebrospinal fluid barrier (BCSFB), a contribution by the blood-brain barrier (BBB), which serves as a major portal for protein delivery to brain, cannot be ruled out. In this study, we assessed the permeability-surface area product (PS) of radioiodinated ApoA-I (125I-ApoA-I) in various brain regions of wild-type rats after an intravenous bolus injection. The PS value at the cortex, caudate putamen, hippocampus, thalamus, brain stem, and cerebellum was found to be 0.39, 0.28, 0.28, 0.36, 0.69, and 0.76 (ml/g per second × 10-6), respectively. Solutes delivered into brain via the BCSFB are expected to show greater accumulation in the thalamus due to its periventricular location. The modest permeability for 125I-ApoA-I into the thalamus relative to other regions suggests that BCSFB transport accounts for only a portion of total brain uptake and thus BBB transport cannot be ruled out. In addition, we show that Alexa Flour 647-labeled ApoA-I (AF647-ApoA-I) undergoes clathrin-independent and cholesterol-mediated endocytosis in transformed human cerebral microvascular endothelial cells (hCMEC/D3). Further, Z-series confocal images of the hCMEC/D3 monolayers and Western blot detection of intact ApoA-I on the abluminal side demonstrated AF647-ApoA-I transcytosis across the endothelium. These findings implicate the BBB as a significant portal for ApoA-I delivery into brain.


Assuntos
Apolipoproteína A-I/metabolismo , Barreira Hematoencefálica/metabolismo , Colesterol/metabolismo , Clatrina/metabolismo , Endocitose , Animais , Apolipoproteína A-I/sangue , Barreira Hematoencefálica/citologia , Endotélio/metabolismo , Humanos , Masculino , Permeabilidade , Transporte Proteico , Ratos , Ratos Sprague-Dawley
8.
Nanomedicine ; 16: 258-266, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30300748

RESUMO

Accumulation of amyloid beta (Aß) peptides in the cerebral vasculature, referred to as cerebral amyloid angiopathy (CAA), is widely observed in Alzheimer's disease (AD) brain and was shown to accelerate cognitive decline. There is no effective method for detecting cerebrovascular amyloid (CVA) and treat CAA. The targeted nanoparticles developed in this study effectively migrated from the blood flow to the vascular endothelium as determined by using quartz crystal microbalance with dissipation monitoring (QCM-D) technology. We also improved the stability, and blood-brain barrier (BBB) transcytosis of targeted nanoparticles by coating them with a cationic BBB penetrating peptide (K16ApoE). The K16ApoE-Targeted nanoparticles demonstrated specific targeting of vasculotropic DutchAß40 peptide accumulated in the cerebral vasculature. Moreover, K16ApoE-Targeted nanoparticles demonstrated significantly greater uptake into brain and provided specific MRI contrast to detect brain amyloid plaques.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Imageamento por Ressonância Magnética/métodos , Nanopartículas/química , Animais , Barreira Hematoencefálica/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Cães , Humanos , Células Madin Darby de Rim Canino
9.
J Cereb Blood Flow Metab ; 38(5): 904-918, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28569090

RESUMO

Impaired brain clearance of amyloid-beta peptides (Aß) 40 and 42 across the blood-brain barrier (BBB) is believed to be one of the pathways responsible for Alzheimer's disease (AD) pathogenesis. Hyperinsulinemia prevalent in type II diabetes was shown to damage cerebral vasculature and increase Aß accumulation in AD brain. However, there is no clarity on how aberrations in peripheral insulin levels affect Aß accumulation in the brain. This study describes, for the first time, an intricate relation between plasma insulin and Aß transport at the BBB. Upon peripheral insulin administration in wild-type mice: the plasma clearance of Aß40 increased, but Aß42 clearance reduced; the plasma-to-brain influx of Aß40 increased, and that of Aß42 reduced; and the clearance of intracerebrally injected Aß40 decreased, whereas Aß42 clearance increased. In hCMEC/D3 monolayers (in vitro BBB model) exposed to insulin, the luminal uptake and luminal-to-abluminal permeability of Aß40 increased and that of Aß42 reduced; the abluminal-to-luminal permeability of Aß40 decreased, whereas Aß42 permeability increased. Moreover, Aß cellular trafficking machinery was altered. In summary, Aß40 and Aß42 demonstrated distinct distribution kinetics in plasma and brain compartments, and insulin differentially modulated their distribution. Cerebrovascular disease and metabolic disorders may disrupt this intricate homeostasis and aggravate AD pathology.


Assuntos
Peptídeos beta-Amiloides/farmacocinética , Química Encefálica/efeitos dos fármacos , Insulina/farmacologia , Doença de Alzheimer , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/sangue , Animais , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Humanos , Camundongos , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/farmacocinética , Transporte Proteico , Distribuição Tecidual/efeitos dos fármacos
10.
J Gerontol A Biol Sci Med Sci ; 72(6): 780-785, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27516624

RESUMO

Osteoarthritis (OA) is the leading form of arthritis in the elderly, causing pain, disability, and immobility. OA has been associated with accumulation of senescent cells in or near joints. However, evidence for a causal link between OA and cellular senescence is lacking. Here, we present a novel senescent cell transplantation model involving injection of small numbers of senescent or nonsenescent cells from the ear cartilage of luciferase-expressing mice into the knee joint area of wild-type mice. By using bioluminescence and 18FDG PET imaging, we could track the injected cells in vivo for more than 10 days. Transplanting senescent cells into the knee region caused leg pain, impaired mobility, and radiographic and histological changes suggestive of OA. Transplanting nonsenescent cells had less of these effects. Thus, senescent cells can induce an OA-like state and targeting senescent cells could be a promising strategy for treating OA.


Assuntos
Senescência Celular , Fibroblastos/transplante , Osteoartrite/etiologia , Joelho de Quadrúpedes , Animais , Fibroblastos/efeitos da radiação , Fluordesoxiglucose F18 , Injeções Intra-Articulares , Medições Luminescentes , Camundongos Endogâmicos C57BL , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Joelho de Quadrúpedes/diagnóstico por imagem , beta-Galactosidase/metabolismo
11.
PLoS One ; 9(5): e97655, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24847943

RESUMO

BACKGROUND: Rapid pre-clinical evaluation of chemotherapeutic agents against brain cancers and other neurological disorders remains largely unattained due to the presence of the blood-brain barrier (BBB), which limits transport of most therapeutic compounds to the brain. A synthetic peptide carrier, K16ApoE, was previously developed that enabled transport of target proteins to the brain by mimicking a ligand-receptor system. The peptide carrier was found to generate transient BBB permeability, which was utilized for non-covalent delivery of cisplatin, methotrexate and other compounds to the brain. APPROACH: Brain delivery of the chemotherapeutics and other agents was achieved either by injecting the carrier peptide and the drugs separately or as a mixture, to the femoral vein. A modification of the method comprised injection of K16ApoE pre-mixed with cetuximab, followed by injection of a 'small-molecule' drug. PRINCIPAL FINDINGS: Seven-of-seven different small molecules were successfully delivered to the brain via K16ApoE. Depending on the method, brain uptake with K16ApoE was 0.72-1.1% for cisplatin and 0.58-0.92% for methotrexate (34-50-fold and 54-92 fold greater for cisplatin and methotrexate, respectively, with K16ApoE than without). Visually intense brain-uptake of Evans Blue, Light Green SF and Crocein scarlet was also achieved. Direct intracranial injection of EB show locally restricted distribution of the dye in the brain, whereas K16ApoE-mediated intravenous injection of EB resulted in the distribution of the dye throughout the brain. Experiments with insulin suggest that ligand-receptor signaling intrinsic to the BBB provides a natural means for passive transport of some molecules across the BBB. SIGNIFICANCE: The results suggest that the carrier peptide can non-covalently transport various chemotherapeutic agents to the brain. Thus, the method offers an avenue for pre-clinical evaluation of various small and large therapeutic molecules against brain tumors and other neurological disorders.


Assuntos
Barreira Hematoencefálica/metabolismo , Cisplatino/administração & dosagem , Cisplatino/metabolismo , Portadores de Fármacos/metabolismo , Metotrexato/administração & dosagem , Metotrexato/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Corantes/metabolismo , Portadores de Fármacos/química , Feminino , Injeções Intravenosas , Insulina/farmacologia , Radioisótopos do Iodo/metabolismo , Camundongos , Dados de Sequência Molecular , Peptídeos/química
12.
J Control Release ; 185: 121-9, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24735640

RESUMO

Cerebral amyloid angiopathy (CAA) is characterized by the deposition of amyloid beta (Aß) proteins within the walls of the cerebral vasculature with subsequent aggressive vascular inflammation leading to recurrent hemorrhagic strokes. The objective of the study was to develop theranostic nanovehicles (TNVs) capable of a) targeting cerebrovascular amyloid; b) providing magnetic resonance imaging (MRI) contrast for the early detection of CAA; and c) treating cerebrovascular inflammation resulting from CAA. The TNVs comprised of a polymeric nanocore made from Magnevist (MRI contrast agent) conjugated chitosan. The nanocore was also loaded with cyclophosphamide (CYC), an immunosuppressant shown to reduce the cerebrovascular inflammation in CAA. Putrescine modified F(ab')2 fragment of anti-amyloid antibody, IgG4.1 (pF(ab')24.1) was conjugated to the surface of the nanocore to target cerebrovascular amyloid. The average size of the control chitosan nanoparticles (conjugated with albumin and are devoid of Magnevist, CYC, and pF(ab')24.1) was 164±1.2 nm and that of the TNVs was 239±4.1 nm. The zeta potential values of the CCNs and TNVs were 21.6±1.7 mV and 11.9±0.5 mV, respectively. The leakage of Magnevist from the TNVs was a modest 0.2% over 4 days, and the CYC release from the TNVs followed Higuchi's model that describes sustained drug release from polymeric matrices. The studies conducted in polarized human microvascular endothelial cell monolayers (hCMEC/D3) in vitro as well as in mice in vivo have demonstrated the ability of TNVs to target cerebrovascular amyloid. In addition, the TNVs provided contrast for imaging cerebrovascular amyloid using MRI and single photon emission computed tomography. Moreover, the TNVs were shown to reduce pro-inflammatory cytokine production by the Aß challenged blood brain barrier (BBB) endothelium more effectively than the cyclophosphamide alone.


Assuntos
Angiopatia Amiloide Cerebral/diagnóstico , Sistemas de Liberação de Medicamentos , Nanopartículas/uso terapêutico , Nanoestruturas/uso terapêutico , Placa Amiloide/patologia , Peptídeos beta-Amiloides , Animais , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Angiopatia Amiloide Cerebral/terapia , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Gadolínio/administração & dosagem , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Inflamação/diagnóstico , Inflamação/terapia , Imageamento por Ressonância Magnética , Camundongos , Nanopartículas/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único
13.
Biomaterials ; 35(6): 1967-76, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24331706

RESUMO

Cerebral amyloid angiopathy (CAA) results from the accumulation of Aß proteins primarily within the media and adventitia of small arteries and capillaries of the cortex and leptomeninges. CAA affects a majority of Alzheimer's disease (AD) patients and is associated with a rapid decline in cognitive reserve. Unfortunately, there is no pre-mortem diagnosis available for CAA. Furthermore, treatment options are few and relatively ineffective. To combat this issue, we have designed nanovehicles (nanoparticles-IgG4.1) capable of targeting cerebrovascular amyloid (CVA) and serving as early diagnostic and therapeutic agents. These nanovehicles were loaded with Gadolinium (Gd) based (Magnevist(®)) magnetic resonance imaging contrast agents or single photon emission computed tomography (SPECT) agents, such as (125)I. In addition, the nanovehicles carry either anti-inflammatory and anti-amyloidogenic agents such as curcumin or immunosuppressants such as dexamethasone, which were previously shown to reduce cerebrovascular inflammation. Owing to the anti-amyloid antibody (IgG4.1) grafted on the surface, the nanovehicles are capable of specifically targeting CVA deposits. The nanovehicles effectively marginate from the blood flow to the vascular wall as determined by using quartz crystal microbalance with dissipation monitoring (QCM-D) technology. They demonstrate excellent distribution to the brain vasculature and target CVA, thus providing MRI and SPECT contrast specific to the CVA in the brain. In addition, they also display the potential to carry therapeutic agents to reduce cerebrovascular inflammation associated with CAA, which is believed to trigger hemorrhage in CAA patients.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Encéfalo/patologia , Animais , Encéfalo/metabolismo , Linhagem Celular , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/terapia , Quitosana/metabolismo , Humanos , Imageamento por Ressonância Magnética , Camundongos , Placa Amiloide , Tomografia Computadorizada de Emissão de Fóton Único
14.
Mol Pharm ; 10(5): 1557-65, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23249146

RESUMO

Amyloid-ß (Aß) deposition in the brain vasculature results in cerebral amyloid angiopathy (CAA), which occurs in about 80% of Alzheimer's disease (AD) patients. While Aß42 predominates parenchymal amyloid plaques in AD brain, Aß40 is prevalent in the cerebrovascular amyloid. Dutch mutation of Aß40 (E22Q) promotes aggressive cerebrovascular accumulation and leads to severe CAA in the mutation carriers; knowledge of how DutchAß40 drives this process more efficiently than Aß40 could reveal various pathophysiological events that promote CAA. In this study we have demonstrated that DutchAß40 shows preferential accumulation in the blood-brain-barrier (BBB) endothelial cells due to its inefficient blood-to-brain transcytosis. Consequently, DutchAß40 establishes a permeation barrier in the BBB endothelium, prevents its own clearance from the brain, and promotes the formation of amyloid deposits in the cerebral microvessels. The BBB endothelial accumulation of native Aß40 is not robust enough to exercise such a significant impact on its brain clearance. Hence, the cerebrovascular accumulation of Aß40 is slow and may require other copathologies to precipitate into CAA. In conclusion, the magnitude of Aß accumulation in the BBB endothelial cells is a critical factor that promotes CAA; hence, clearing vascular endothelium of Aß proteins may halt or even reverse CAA.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/genética , Animais , Bovinos , Células Cultivadas , Angiopatia Amiloide Cerebral Familiar/genética , Angiopatia Amiloide Cerebral Familiar/metabolismo , Células Endoteliais/metabolismo , Humanos , Modelos Neurológicos , Proteínas Mutantes/sangue , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transporte Proteico , Transcitose
16.
PLoS One ; 6(6): e20347, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738572

RESUMO

BACKGROUND: Pancreatic cancer is the fourth leading cause of cancer related deaths in America. Monoclonal antibodies are a viable treatment option for inhibiting cancer growth. Tumor specific drug delivery could be achieved utilizing these monoclonal antibodies as targeting agents. This type of designer therapeutic is evolving and with the use of gold nanoparticles it is a promising approach to selectively deliver chemotherapeutics to malignant cells. Gold nanoparticles (GNPs) are showing extreme promise in current medicinal research. GNPs have been shown to non-invasively kill tumor cells by hyperthermia using radiofrequency. They have also been implemented as early detection agents due to their unique X-ray contrast properties; success was revealed with clear delineation of blood capillaries in a preclinical model by CT (computer tomography). The fundamental parameters for intelligent design of nanoconjugates are on the forefront. The goal of this study is to define the necessary design parameters to successfully target pancreatic cancer cells. METHODOLOGY/PRINCIPAL FINDINGS: The nanoconjugates described in this study were characterized with various physico-chemical techniques. We demonstrate that the number of cetuximab molecules (targeting agent) on a GNP, the hydrodynamic size of the nanoconjugates, available reactive surface area and the ability of the nanoconjugates to sequester EGFR (epidermal growth factor receptor), all play critical roles in effectively targeting tumor cells in vitro and in vivo in an orthotopic model of pancreatic cancer. CONCLUSION: Our results suggest the specific targeting of tumor cells depends on a number of crucial components 1) targeting agent to nanoparticle ratio 2) availability of reactive surface area on the nanoparticle 3) ability of the nanoconjugate to bind the target and 4) hydrodynamic diameter of the nanoconjugate. We believe this study will help define the design parameters for formulating better strategies for specifically targeting tumors with nanoparticle conjugates.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ouro/química , Nanopartículas Metálicas/química , Nanoconjugados/química , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Linhagem Celular Tumoral , Cetuximab , Humanos , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Ensaios Antitumorais Modelo de Xenoenxerto
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA