Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
FASEB J ; 28(6): 2620-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24604080

ABSTRACT

Tau alterations are now considered an executor of neuronal demise and cognitive dysfunction in Alzheimer's disease (AD). Mouse models combining amyloidosis and tauopathy and their parental counterparts are important tools to further investigate the interplay of abnormal amyloid-ß (Aß) and Tau species in pathogenesis, synaptic and neuronal dysfunction, and cognitive decline. Here, we crossed APP/PS1 mice with 5 early-onset familial AD mutations (5xFAD) and TauP301S (PS19) transgenic mice, denoted F(+)/T(+) mice, and phenotypically compared them to their respective parental strains, denoted F(+)/T(-) and F(-)/T(+) respectively, as controls. We found dramatically aggravated tauopathy (~10-fold) in F(+)/T(+) mice compared to the parental F(-)/T(+) mice. In contrast, amyloidosis was unaltered compared to the parental F(+)/T(-) mice. Tauopathy was invariably and very robustly aggravated in hippocampal and cortical brain regions. Most important, F(+)/T(+) displayed aggravated cognitive deficits in a hippocampus-dependent spatial navigation task, compared to the parental F(+)/T(-) strain, while parental F(-)/T(+) mice did not display cognitive impairment. Basal synaptic transmission was impaired in F(+)/T(+) mice compared to nontransgenic mice and the parental strains (≥40%). Finally, F(+)/T(+) mice displayed a significant hippocampal atrophy (~20%) compared to nontransgenic mice, in contrast to the parental strains. Our data indicate for the first time that pathological Aß species (or APP/PS1) induced changes in Tau contribute to cognitive deficits correlating with synaptic deficits and hippocampal atrophy in an AD model. Our data lend support to the amyloid cascade hypothesis with a role of pathological Aß species as initiator and pathological Tau species as executor.


Subject(s)
Alzheimer Disease/pathology , Cognition Disorders/etiology , Synaptic Transmission , Tauopathies/complications , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Atrophy/pathology , Cognition Disorders/pathology , Disease Models, Animal , Female , Glycogen Synthase Kinase 3/metabolism , Hippocampus/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Maze Learning , Mice , Mice, Transgenic , Presenilin-1/genetics , Tauopathies/pathology , tau Proteins/genetics
2.
Neurobiol Dis ; 62: 100-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24076100

ABSTRACT

Neurofibrillary degeneration in transgenic models of tauopathies has been observed to be enhanced when these models are crossed with transgenic models developing an Aß pathology. The mechanisms leading to this enhanced tau pathology are not well understood. We have performed a detailed analysis of tau misprocessing in a new transgenic mouse model combining APP, PS1 and tau mutations (5xFAD×Tg30 mice) by comparison with littermates expressing only a FTD mutant tau (Tg30 mice). These 5xFAD×Tg30 mice showed a more severe deficient motor phenotype than Tg30 mice and developed with age a dramatically accelerated NFT load in the brain compared to Tg30 mice. Insoluble tau in 5xFAD×Tg30 mice compared to insoluble tau in Tg30 mice showed increased phosphorylation, enhanced misfolding and truncation changes mimicking more closely the post-translational changes characteristic of PHF-tau in Alzheimer's disease. Endogenous wild-type mouse tau was recruited at much higher levels in insoluble tau in 5xFAD×Tg30 than in Tg30 mice. Extracellular amyloid load, Aß40 and Aß42, ß-CTFs and ß-CTF phosphorylation levels were lower in 5xFAD×Tg30 mice than in 5xFAD mice. Despite this reduction of Aß, a significant hippocampal neuronal loss was observed in 5xFAD×Tg30 but not in 5xFAD mice indicating its closer association with increased tau pathology. This 5xFAD×Tg30 model thus mimics more faithfully tau pathology and neuronal loss observed in AD and suggests that additional post-translational changes in tau and self-recruitment of endogenous tau drive the enhanced tau pathology developing in the presence of Aß pathology.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cerebral Cortex/ultrastructure , Plaque, Amyloid/ultrastructure , Presenilin-1/genetics , tau Proteins/genetics , tau Proteins/metabolism , Age Factors , Amyloid beta-Protein Precursor/metabolism , Animals , Hippocampus/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Phosphorylation , Presenilin-1/metabolism , Protein Folding , Pyramidal Cells/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Survival Rate , tau Proteins/chemistry
3.
Langmuir ; 29(5): 1498-509, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23305497

ABSTRACT

Antibody microarrays are powerful and high-throughput tools for screening and identifying tumor markers from small sample volumes of only a few microliters. Optimization of surface chemistry and spotting conditions are crucial parameters to enhance antibodies' immobilization efficiency and to maintain their biological activity. Here, we report the implementation of an antibody microarray for the detection of tumor markers involved in colorectal cancer. Three-dimensional microstructured glass slides were functionalized with three different aminated molecules ((3-aminopropyl)dimethylethoxysilane (APDMES), Jeffamine, and chitosan) varying in their chain length, their amine density, and their hydrophilic/hydrophobic balance. The physicochemical properties of the resulting surfaces were characterized. Antibody immobilization efficiency through physical interaction was studied as a function of surface properties as well as a function of the immobilization conditions. The results show that surface energy, steric hindrance, and pH of spotting buffer have great effects on protein immobilization. Under optimal conditions, biological activities of four immobilized antitumor marker antibodies were evaluated in multiplex immunoassay for the detection of the corresponding tumor markers. Results indicated that the chitosan functionalized surface displayed the highest binding capacity and allowed to retain maximal biological activity of the four tested antibody/antigen systems. Thus, we successfully demonstrated the application of amino-based surface modification for antibody microarrays to efficiently detect tumor markers.


Subject(s)
Amines/chemistry , Antibodies/chemistry , Biomarkers, Tumor/analysis , Chitosan/chemistry , Colorectal Neoplasms/diagnosis , Propylamines/chemistry , Silanes/chemistry , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Protein Array Analysis , Surface Properties
4.
Am J Pathol ; 181(6): 1928-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23026200

ABSTRACT

Lack of tau expression has been reported to protect against excitotoxicity and to prevent memory deficits in mice expressing mutant amyloid precursor protein (APP) identified in familial Alzheimer disease. In APP mice, mutant presenilin 1 (PS1) enhances generation of Aß42 and inhibits cell survival pathways. It is unknown whether the deficient phenotype induced by concomitant expression of mutant PS1 is rescued by absence of tau. In this study, we have analyzed the effect of tau deletion in mice expressing mutant APP and PS1. Although APP/PS1/tau(+/+) mice had a reduced survival, developed spatial memory deficits at 6 months and motor impairments at 12 months, these deficits were rescued in APP/PS1/tau(-/-) mice. Neuronal loss and synaptic loss in APP/PS1/tau(+/+) mice were rescued in the APP/PS1/tau(-/-) mice. The amyloid plaque burden was decreased by roughly 50% in the cortex and the spinal cord of the APP/PS1/tau(-/-) mice. The levels of soluble and insoluble Aß40 and Aß42, and the Aß42/Aß40 ratio were reduced in APP/PS1/tau(-/-) mice. Levels of phosphorylated APP, of ß-C-terminal fragments (CTFs), and of ß-secretase 1 (BACE1) were also reduced, suggesting that ß-secretase cleavage of APP was reduced in APP/PS1/tau(-/-) mice. Our results indicate that tau deletion had a protective effect against amyloid induced toxicity even in the presence of mutant PS1 and reduced the production of Aß.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Neurons/pathology , Presenilin-1/metabolism , Protein Processing, Post-Translational , tau Proteins/deficiency , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases/metabolism , Cell Death , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Humans , Memory, Short-Term , Mice , Mice, Transgenic , Motor Activity , Neuroglia/metabolism , Neuroglia/pathology , Neurons/ultrastructure , Phosphorylation , Phosphothreonine/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Solubility , Survival Analysis , Synapses/pathology , Synapses/ultrastructure , tau Proteins/metabolism
5.
Nano Lett ; 12(5): 2573-8, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22458647

ABSTRACT

The development of low-temperature carbonization procedures promises to provide novel nanostructured carbon materials that are of high current interest in materials science and technology. Here, we report a "wet-chemical" carbonization method that utilizes hexayne amphiphiles as metastable carbon precursors. Nearly perfect control of the nanoscopic morphology was achieved by self-assembly of the precursors into colloidal aggregates with tailored diameter in water. Subsequent carbonization furnished carbon nanocapsules with a carbon microstructure resembling graphite-like amorphous carbon materials.

6.
ACS Appl Mater Interfaces ; 4(2): 573-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22257109

ABSTRACT

Different anchoring groups have been studied with the aim of covalently binding organic linkers to the surface of alumina ceramic foams. The results suggested that a higher degree of functionalization was achieved with a pyrogallol derivative--as compared to its catechol analogue--based on the XPS analysis of the ceramic surface. The conjugation of organic ligands to the surface of these alumina materials was corroborated by DNP-MAS NMR measurements.

7.
Nat Mater ; 10(6): 456-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21552270

ABSTRACT

A clean and efficient way to overcome the limited supply of fossil fuels and the greenhouse effect is the production of hydrogen fuel from sunlight and water through the semiconductor/water junction of a photoelectrochemical cell, where energy collection and water electrolysis are combined into a single semiconductor electrode. We present a highly active photocathode for solar H(2) production, consisting of electrodeposited cuprous oxide, which was protected against photocathodic decomposition in water by nanolayers of Al-doped zinc oxide and titanium oxide and activated for hydrogen evolution with electrodeposited Pt nanoparticles. The roles of the different surface protection components were investigated, and in the best case electrodes showed photocurrents of up to -7.6 mA cm(-2) at a potential of 0 V versus the reversible hydrogen electrode at mild pH. The electrodes remained active after 1 h of testing, cuprous oxide was found to be stable during the water reduction reaction and the Faradaic efficiency was estimated to be close to 100%.

8.
Fundam Clin Pharmacol ; 25(1): 115-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20199584

ABSTRACT

Microglia are the professional phagocytes of the brain and express phagocytic receptors such as complement receptor 3 (CR3 or CD11b/CD18). Using mimics of the amyloid deposit made of heat-killed yeasts coated with either Aß 1-40 or Aß 1-42, we were able to study how microglia interacted with and ingested these particles in vitro. We have shown previously that the low density lipoprotein receptor-related protein (LRP) is largely implied in the phagocytosis of Aß 1-42-opsonized heat-killed yeasts and partly in that of Aß 1-40-opsonized heat-killed yeasts. Here, we report that antibodies against CD11b or CD18 reduced the uptake of the artificial amyloid deposit by microglial cell showing that CR3 is involved in the mechanism. Moreover, a concomitant inhibition of LRP and CR3 completely blocked the ingestion of both kinds of particles suggesting that no other receptors participate to this mechanism.


Subject(s)
CD11b Antigen/immunology , CD18 Antigens/immunology , Macrophage-1 Antigen/metabolism , Microglia/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line , Laminaria/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Macrophage-1 Antigen/immunology , Mice , Mice, Inbred BALB C , Microglia/immunology , Peptide Fragments/metabolism , Saccharomyces cerevisiae/metabolism
9.
Mol Med ; 17(3-4): 149-62, 2011.
Article in English | MEDLINE | ID: mdl-21170472

ABSTRACT

Several large population-based or clinical trial studies have suggested that certain dihydropyridine (DHP) L-type calcium channel blockers (CCBs) used for the treatment of hypertension may confer protection against the development of Alzheimer disease (AD). However, other studies with drugs of the same class have shown no beneficial clinical effects. To determine whether certain DHPs are able to impact underlying disease processes in AD (specifically the accumulation of the Alzheimer Aß peptide), we investigated the effect of several antihypertensive DHPs and non-DHP CCBs on Aß production. Among the antihypertensive DHPs tested, a few, including nilvadipine, nitrendipine and amlodipine inhibited Aß production in vitro, whereas others had no effect or raised Aß levels. In vivo, nilvadipine and nitrendipine acutely reduced brain Aß levels in a transgenic mouse model of AD (Tg PS1/APPsw) and improved Aß clearance across the blood-brain barrier (BBB), whereas amlodipine and nifedipine were ineffective showing that the Aß-lowering activity of the DHPs is independent of their antihypertensive activity. Chronic oral treatment with nilvadipine decreased Aß burden in the brains of Tg APPsw (Tg2576) and Tg PS1/APPsw mice, and also improved learning abilities and spatial memory. Our data suggest that the clinical benefit conferred by certain antihypertensive DHPs against AD is unrelated to their antihypertensive activity, but rely on their ability to lower brain Aß accumulation by affecting both Aß production and Aß clearance across the BBB.


Subject(s)
Amyloid beta-Peptides/metabolism , Antihypertensive Agents/pharmacology , Blood-Brain Barrier/drug effects , Dihydropyridines/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Amlodipine/pharmacology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/physiopathology , CHO Cells , Calcium Channel Blockers/pharmacology , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred Strains , Mice, Transgenic , Nifedipine/analogs & derivatives , Nifedipine/pharmacology , Nitrendipine/pharmacology
10.
J Neurosci ; 30(34): 11251-8, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20739545

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia among the aging population and is characterized pathologically by the progressive intracerebral accumulation of beta-amyloid (Abeta) peptides and neurofibrillary tangles. The level of proangiogenic growth factors and inflammatory mediators with proangiogenic activity is known to be elevated in AD brains which has led to the supposition that the cerebrovasculature of AD patients is in a proangiogenic state. However, angiogenesis depends on the balance between proangiogenic and antiangiogenic factors and the brains of AD patients also show an accumulation of endostatin and Abeta peptides which have been shown to be antiangiogenic. To determine whether angiogenesis is compromised in the brains of two transgenic mouse models of AD overproducing Abeta peptides (Tg APPsw and Tg PS1/APPsw mice), we assessed the growth and vascularization of orthotopically implanted murine gliomas since they require a high degree of angiogenesis to sustain their growth. Our data reveal that intracranial tumor growth and angiogenesis is significantly reduced in Tg APPsw and Tg PS1/APPsw mice compared with their wild-type littermates. In addition, we show that Abeta inhibits the angiogenesis stimulated by glioma cells when cocultured with human brain microvascular cells on a Matrigel layer. Altogether our data suggest that the brain of transgenic mouse models of AD does not constitute a favorable environment to support neoangiogenesis and may explain why vascular insults synergistically precipitate the cognitive presentation of AD.


Subject(s)
Alzheimer Disease/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Glioma/pathology , Neovascularization, Pathologic/pathology , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Animals , Brain Neoplasms/prevention & control , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cricetinae , Disease Models, Animal , Glioma/genetics , Glioma/prevention & control , Humans , Mice , Mice, Transgenic , Neoplasm Transplantation/methods , Neoplasm Transplantation/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control
11.
Int J Pept Res Ther ; 16(1): 23-30, 2010.
Article in English | MEDLINE | ID: mdl-20473341

ABSTRACT

The inhibition of angiogenesis is regarded as a promising avenue for cancer treatment. Although some antiangiogenic compounds are in the process of development and testing, these often prove ineffective in vivo, therefore the search for new inhibitors is critical. We have recently identified a ten amino acid fragment of the Alzheimer Abeta peptide that is anti-angiogenic both in vitro and in vivo. In the present study, we investigated the antitumoral potential of this decapeptide using human MCF-7 breast carcinoma xenografts nude mice. We observed that this decapeptide was able to suppress MCF-7 tumor growth more potently than the antiestrogen tamoxifen. Inhibition of tumor vascularization as determined by PECAM-1 immunostaining and decreased tumor cell proliferation as determined by Ki67 immunostaining were observed following treatment with the Abeta fragment. In vitro, this peptide had no direct impact on MCF-7 tumor cell proliferation and survival suggesting that the inhibition of tumor growth and tumor cell proliferation observed in vivo is related to the antiangiogenic activity of the peptide. Taken together these data suggest that this short Abeta derivative peptide may constitute a new antitumoral agent.

12.
J Neuroinflammation ; 7: 17, 2010 Mar 08.
Article in English | MEDLINE | ID: mdl-20211007

ABSTRACT

BACKGROUND: Abeta deposits represent a neuropathological hallmark of Alzheimer's disease (AD). Both soluble and insoluble Abeta species are considered to be responsible for initiating the pathological cascade that eventually leads to AD. Therefore, the identification of therapeutic approaches that can lower Abeta production or accumulation remains a priority. NFkappaB has been shown to regulate BACE-1 expression level, the rate limiting enzyme responsible for the production of Abeta. We therefore explored whether the known NFkappaB inhibitor celastrol could represent a suitable compound for decreasing Abeta production and accumulation in vivo. METHODS: The effect of celastrol on amyloid precursor protein (APP) processing, Abeta production and NFkappaB activation was investigated by western blotting and ELISAs using a cell line overexpressing APP. The impact of celastrol on brain Abeta accumulation was tested in a transgenic mouse model of AD overexpressing the human APP695sw mutation and the presenilin-1 mutation M146L (Tg PS1/APPsw) by immunostaining and ELISAs. An acute treatment with celastrol was investigated by administering celastrol intraperitoneally at a dosage of 1 mg/Kg in 35 week-old Tg PS1/APPsw for 4 consecutive days. In addition, a chronic treatment (32 days) with celastrol was tested using a matrix-driven delivery pellet system implanted subcutaneously in 5 month-old Tg PS1/APPsw to ensure a continuous daily release of 2.5 mg/Kg of celastrol. RESULTS: In vitro, celastrol dose dependently prevented NFkappaB activation and inhibited BACE-1 expression. Celastrol potently inhibited Abeta1-40 and Abeta1-42 production by reducing the beta-cleavage of APP, leading to decreased levels of APP-CTFbeta and APPsbeta. In vivo, celastrol appeared to reduce the levels of both soluble and insoluble Abeta1-38, Abeta1-40 and Abeta1-42. In addition, a reduction in Abeta plaque burden and microglial activation was observed in the brains of Tg PS1/APPsw following a chronic administration of celastrol. CONCLUSIONS: Overall our data suggest that celastrol is a potent Abeta lowering compound that acts as an indirect BACE-1 inhibitor possibly by regulating BACE-1 expression level via an NFkappaB dependent mechanism. Additional work is required to determine whether chronic administration of celastrol can be safely achieved with cognitive benefits in a transgenic mouse model of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Triterpenes/therapeutic use , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Carcinogens/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Pentacyclic Triterpenes , Phorbol Esters/pharmacology , Presenilin-1/genetics , Signal Transduction/drug effects , Transfection/methods , Triterpenes/chemistry
13.
J Neurochem ; 112(1): 66-76, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19818105

ABSTRACT

Beta-amyloid peptides (Abeta) are the major constituents of senile plaques and cerebrovascular deposits in the brains of Alzheimer's disease patients. We have shown previously that soluble forms of Abeta are anti-angiogenic both in vitro and in vivo. However, the mechanism of the anti-angiogenic activity of Abeta peptides is unclear. In this study, we examined the effects of Abeta1-42 on vascular endothelial growth factor receptor 2 (VEGFR-2) signaling, which plays a key role in angiogenesis. Abeta inhibited VEGF-induced migration of endothelial cells, as well as VEGF-induced permeability of an in vitro model of the blood brain barrier. Consistently, exogenous VEGF dose-dependently antagonized the anti-angiogenic activity of Abeta in a capillary network assay. Abeta1-42 also blocked VEGF-induced tyrosine phosphorylation of VEGFR-2 in two types of primary endothelial cells, suggesting an antagonistic action of Abeta toward VEGFR-2 signaling in cells. Moreover, Abeta was able to directly interact with the extracellular domain of VEGFR-2 and to compete with the binding of VEGF to its receptor in a cell-free assay. Co-immunoprecipitation experiments confirmed that Abeta can bind VEGFR-2 both in vitro and in vivo. Altogether, our data suggest that Abeta acts as an antagonist of VEGFR-2 and provide a mechanism explaining the anti-angiogenic activity of Abeta peptides.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Peptide Fragments/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Angiogenesis Inhibitors/antagonists & inhibitors , Angiogenesis Inhibitors/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Mice , Mice, Transgenic , Peptide Fragments/metabolism , Protein Binding/physiology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
14.
ACS Chem Biol ; 3(12): 777-89, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19067586

ABSTRACT

Alzheimer's disease (AD) is a progressive chronic disorder that leads to cognitive decline. Several studies have associated up-regulation of some of the chemokines and/or their receptors with altered APP processing leading to increased production of beta-amyloid protein (Abeta) and AD pathological changes. However, there is no direct evidence to date to determine whether the altered processing of APP results in up-regulation of these receptors or whether the up-regulation of the chemokine receptors causes modulated processing of APP. In the current study, we demonstrate that treatment of the chemokine receptor CXCR2 with agonists leads to enhancement of Abeta production and treatment with antagonists or immunodepletion of CXCR2's endogenous agonists leads to Abeta inhibition. Further, we found that the inhibitory effect of the antagonist of CXCR2 on Abeta40 and Abeta42 is mediated via gamma-secretase, specifically through reduction in expression of presenilin (PS), one of the gamma-secretase components. Also, in vivo chronic treatment with a CXCR2 antagonist blocked Abeta40 and Abeta42 production. Using small interfering RNAs for CXCR2, we further showed that knockdown of CXCR2 in vitro accumulates gamma-secretase substrates C99 and C83 with reduced production of both Abeta40 and Abeta42. Taken together, these findings strongly suggest for the first time that up-regulation of the CXCR2 receptor can be the driving force in increased production of Abeta. Our findings unravel new mechanisms involving the CXCR2 receptor in the pathogenesis of AD and pose it as a potential target for developing novel therapeutics for intervention in this disease. Also, we propose here a new chemical series of interest that can serve as a prototype for drug development.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Receptors, Interleukin-8B/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Animals , CHO Cells , Cells, Cultured , Chemokine CXCL1/pharmacology , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Interleukin-8/pharmacology , Mice , Mice, Transgenic , Phenylurea Compounds/pharmacology , Presenilins/metabolism , RNA, Small Interfering/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Recombinant Proteins/pharmacology , Sulfonamides/pharmacology , Triglycerides/pharmacology , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/pharmacology
15.
Brain Res ; 1231: 132-42, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18606155

ABSTRACT

Neuritic dystrophy with amyloid burden and neurofibrillary tangles are pathological hallmarks of Alzheimer's disease. Genetic disruption of CD40 or CD40L alleviates amyloid burden, astrocytosis, and microgliosis in transgenic animal models of Alzheimer's disease. It has been reported that phosphorylated tau-positive dystrophic neurites are observed in transgenic mice over-expressing human mutant beta-amyloid precursor protein (Tg2576). Here, we studied the pattern of phosphorylated tau (labeled with AT8, CP13, PG5, and PHF1 antibodies) and plaques using immunohistochemical techniques. Phosphorylated tau-positive dystrophic neurites were exclusively associated with Congo red-positive plaques as previously reported. Further, we show that CD40L or CD40 deficiency reduces the mean ratio of dystrophic neurite area to congophilic plaque area and the level of expression of cdk5 and p35/p25 in mice. In addition, we show that in a human neuroblastoma cell line treated with CD40L, cdk5 and p35/p25 are increased. Together, our data suggest that CD40-CD40L interaction has an effect on tau phosphorylation independent of beta-amyloid pathology, and that this effect may occur through a decrease of cdk5 and p35/p25.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Plaque, Amyloid/metabolism , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/immunology , Brain/physiopathology , CD40 Antigens/drug effects , CD40 Antigens/immunology , CD40 Ligand/immunology , CD40 Ligand/pharmacology , Cell Line, Tumor , Chromobox Protein Homolog 5 , Coloring Agents , Congo Red , Cyclin-Dependent Kinase 5/drug effects , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/drug effects , Neurites/immunology , Neurites/metabolism , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/immunology , Neurofibrillary Tangles/metabolism , Phosphorylation/drug effects , Phosphotransferases/drug effects , Phosphotransferases/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/immunology
16.
Amyloid ; 15(1): 5-19, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18266117

ABSTRACT

Abeta peptides are the major constituents of senile plaques and cerebrovascular deposits in the brains of patients with Alzheimer's disease. We have shown previously that Abeta1-40 and Abeta1-42 peptides are potently anti-angiogenic both in vitro and in vivo. The current study characterizes important sequences within the Abeta peptide that are required to exert its anti-angiogenic activity. We have used human umbilical vein endothelial cells to assess the anti-angiogenic activity of short fragments of Abetain vitro in a Matrigel network assay and in vivo in a rat corneal model of angiogenesis. The anti-angiogenic activity of these short peptide fragments is not related to effects on apoptosis or necrosis. Using normal and mutated peptide fragments, we show that the sequence VHHQKLVFF is sufficient to exhibit potent anti-angiogenic effects. This small peptide may therefore have clinical relevance as an anti-angiogenic agent.


Subject(s)
Amyloid beta-Peptides/pharmacology , Angiogenesis Inhibitors/pharmacology , Corneal Neovascularization/metabolism , Endothelial Cells/metabolism , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Umbilical Veins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Motifs/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Brain , Cells, Cultured , Corneal Neovascularization/chemically induced , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Endothelial Cells/pathology , Humans , Mutation , Oligopeptides/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Umbilical Veins/pathology
17.
J Neuroinflammation ; 3: 3, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16504112

ABSTRACT

We have previously shown that transgenic mice carrying a mutant human APP but deficient in CD40L, display a decrease in astrocytosis and microgliosis associated with a lower amount of deposited Abeta. Furthermore, an anti-CD40L treatment causes a diminution of Abeta pathology in the brain and an improved performance in several cognitive tasks in the double transgenic PSAPP mouse model. Although these data suggest a potential role for CD40L in Alzheimer's disease pathology in transgenic mice they do not cast light on whether this effect is due to inhibition of signaling via CD40 or whether it is due to the mitigation of some other unknown role of CD40L. In the present report we have generated APP and PSAPP mouse models with a disrupted CD40 gene and compared the pathological features (such as amyloid burden, astrocytosis and microgliosis that are typical of Alzheimer's disease-like pathology in these transgenic mouse strains) with appropriate controls. We find that all these features are reduced in mouse models deficient for CD40 compared with their littermates where CD40 is present. These data suggest that CD40 signaling is required to allow the full repertoire of AD-like pathology in these mice and that inhibition of the CD40 signaling pathway is a potential therapeutic strategy in Alzheimer's disease.

18.
Fundam Clin Pharmacol ; 20(1): 81-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16448398

ABSTRACT

This study compared three different synthetic reagents (FuGENE 6, Effectene and ExGen 500) for the transfection of human primary myoblasts. We examined the efficiency, cytotoxicity and size of the complexes formed in the presence of different amounts of vector and DNA and with variable amounts of serum. Transfection rates were relatively high for primary cells, especially with FuGENE 6 (20%), which appeared to be the best transfection reagent for these cells, even in the presence of 10% serum. Cultured human myoblasts are an interesting tool for studying neuromuscular diseases and are potentially useful for myoblast transfer therapy studies. Moreover, the efficiency of these transfection reagents in a medium containing 10% serum is promising for possible gene therapy protocols for muscle diseases.


Subject(s)
DNA/metabolism , Indicators and Reagents/chemistry , Lipids/chemistry , Myoblasts, Skeletal/metabolism , Polyethyleneimine/chemistry , Polyethyleneimine/toxicity , Transfection , Cell Culture Techniques , Cell Survival , Cells, Cultured , DNA/chemistry , Humans , Lipids/toxicity , Myoblasts, Skeletal/drug effects , Particle Size , Transfection/methods
19.
Brain Res Mol Brain Res ; 140(1-2): 73-85, 2005 Oct 31.
Article in English | MEDLINE | ID: mdl-16182406

ABSTRACT

Key pathological processes in Alzheimer's disease (AD) include the accumulation of amyloid beta peptide (Abeta) which, in excess, triggers pathological cascades including widespread inflammation, partly reflected by chronic microglial activation. It has previously been suggested that CD40/CD40L interaction promotes AD like pathology in transgenic mice. Thus, amyloid burden, gliosis and hyperphosphorylation of tau are all reduced in transgenic models of AD lacking functional CD40L. We therefore hypothesized that cellular events leading to altered APP metabolism, inflammation and increased tau phosphorylation underlying these observations would be regulated at the genomic level. In the present report, we used the Affymetrix (GeneChip) oligonucleotide microarray U133A to gain insight into the global and simultaneous transcriptomic changes in response to microglia activation after CD40/CD40L ligation. As expected, regulation of elements of the NF-kappaB signaling, chemokine and B cell signaling pathways was observed. Taken together, our data also suggest that CD40 ligation in human microglia specifically perturbs many genes associated with APP processing.


Subject(s)
Alzheimer Disease/genetics , CD40 Antigens/pharmacology , Gene Expression Regulation , Microglia/physiology , Alzheimer Disease/immunology , Amyloid beta-Peptides/genetics , B-Lymphocytes/immunology , CD40 Ligand/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/physiology , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/physiology , Humans , Interleukin-6/physiology , Microglia/immunology , Models, Biological , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Signal Transduction/immunology
20.
Brain Res Mol Brain Res ; 136(1-2): 212-30, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15893605

ABSTRACT

Cerebral amyloid angiopathy is a common pathological feature of patients with Alzheimer's disease (AD) and it is also the hallmark of individuals with a rare autosomal dominant disorder known as hereditary cerebral hemorrhage with amyloidosis-Dutch type. We have shown previously that wild type A(beta) peptides are anti-angiogenic both in vitro and in vivo and could contribute to the compromised cerebrovascular architecture observed in AD. In the present study, we investigated the potential anti-angiogenic activity of the Dutch A(beta)(1-40) (E22Q) peptide. We show that compared to wild type A(beta), freshly solubilized Dutch A(beta) peptide more potently inhibits the formation of capillary structures induced by plating human brain microvascular endothelial cells onto a reconstituted basement membrane. Aggregated/fibrillar preparations of wild type A(beta) and Dutch A(beta) do not appear to be anti-angiogenic in this assay. The stronger anti-angiogenic activity of the Dutch A(beta) compared to wild type A(beta) appears to be related to the increased formation of low molecular weight A(beta) oligomers in the culture medium surrounding human brain microvascular endothelial cells. Using oligonucleotide microarray analysis of human brain microvascular endothelial cells, followed by a genome-scale computational analysis with the Ingenuity Pathways Knowledge Base, networks of genes affected by an anti-angiogenic dose of Dutch A(beta) were identified. This analysis highlights that several biological networks involved in angiogenesis, tumorigenesis, atherosclerosis, cellular migration and proliferation are disrupted in human brain microvascular endothelial cells exposed to Dutch A(beta). Altogether, these data provide new molecular clues regarding the pathological activity of Dutch A(beta) peptide in the cerebrovasculature.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Mutation , Neovascularization, Pathologic/physiopathology , Peptide Fragments/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Blotting, Western/methods , Brain/growth & development , Cell Aggregation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cluster Analysis , Computer Simulation , Congo Red , Dose-Response Relationship, Drug , Endothelium, Vascular/growth & development , Humans , Models, Molecular , Morphogenesis/drug effects , Oligonucleotide Array Sequence Analysis/methods , Peptide Fragments/chemistry , Peptide Fragments/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...