Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Curr Alzheimer Res ; 15(7): 637-642, 2018.
Article in English | MEDLINE | ID: mdl-29637859

ABSTRACT

BACKGROUND: It has been postulated that inadequate clearance of the amyloid ß protein (Aß) plays an important role in the accumulation of Aß in sporadic late onset Alzheimer's disease (AD). While the blood brain barrier (BBB) has taken the center stage in processes involving Aß clearance, little information is available about the role of the lymphatic system. We previously reported that Aß is cleared through the lymphatic system. We now assessed lymphatic Aß clearance by treating a mouse model of AD amyloidosis with melatonin, an Aß aggregation inhibitor and immuno-regulatory neurohormone. OBJECTIVE: To confirm and expand our initial finding that Aß is cleared through the lymphatic system. Lymphatic clearance of metabolic and cellular "waste" products from the brain into the peripheral lymphatic system has been known for a long time. However, except for our prior report, there is no additional experimental data published about Aß being cleared into peripheral lymph nodes. METHODS: For these experiments, we used a transgenic mouse model (Tg2576) that over-expresses a mutant form of the Aß precursor protein (APP) in the brain. We examined levels of Aß in plasma and in lymph nodes of transgenic mice as surrogate markers of vascular and lymphatic clearance, respectively. Aß levels were also measured in the brain and in multiple tissues. RESULTS: Clearance of Aß peptides through the lymphatic system was confirmed in this study. Treatment with melatonin led to the following changes: 1-A statistically significant increase in soluble monomeric Aß40 and an increasing trend in Aß42 in cervical and axillary lymph nodes of treated mice. 2- Statistically significant decreases in oligomeric Aß40 and a decreasing trend Aß42 in the brain. CONCLUSION: The data expands on our prior report that the lymphatic system participates in Aß clearance from the brain. We propose that abnormalities in Aß clearance through the lymphatic system may contribute to the development of cerebral amyloidosis. Melatonin and related indole molecules (i.e., indole- 3-propionic acid) are known to inhibit Aß aggregation although they do not reverse aggregated Aß or amyloid fibrils. Therefore, these substances should be further explored in prevention trials for delaying the onset of cognitive impairment in high risk populations.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloidosis/drug therapy , Lymph Nodes/drug effects , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Amyloidosis/metabolism , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Humans , Lymph Nodes/metabolism , Mice, Transgenic
2.
Indian J Med Res ; 128(4): 545-56, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19106446

ABSTRACT

Exposure to high levels of aluminium (Al) leads to neurofibrillary degeneration and that Al concentration is increased in degenerating neurons in Alzheimer's disease (AD). Nevertheless, the role of Al in AD remains controversial and there is little proof directly interlinking Al to AD. The major problem in understanding Al toxicity is the complex Al speciation chemistry in biological systems. A new dimension is provided to show that Al-maltolate treated aged rabbits can be used as a suitable animal model for understanding the pathology in AD. The intracisternal injection of Al-maltolate into aged New Zealand white rabbits results in pathology that mimics several of the neuropathological, biochemical and behavioural changes as observed in AD. The neurodegenerative effects include the formation of intraneuronal neurofilamentous aggregates that are tau positive, oxidative stress and apoptosis. The present review discusses the role of Al and use of Al-treated aged rabbit as a suitable animal model to understand AD pathogenesis.


Subject(s)
Aluminum/toxicity , Alzheimer Disease/chemically induced , Animals , Disease Models, Animal , Rabbits
3.
Brain Res Rev ; 58(1): 136-48, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18342372

ABSTRACT

DNA is a dynamic molecule, the conformation of which plays a major role in biological function. The non-B-form of DNA conformations are reported in the patho-physiology of diseases like Fragile X-syndrome, Huntington's chorea, Alzheimer's and others. Recently, our laboratory discovered the presence of Z-DNA in the hippocampal region of severely affected Alzheimer's disease (AD) brain samples. Alternate purine-pyrimidine bases, potential sequences adopting Z-DNA, are present in the promoter regions of AD specific genes like amyloid precursor protein (APP), Presenilin and ApoE. Thus, Z-DNA might be involved in the expression of these pathologically important genes. In the present review, we have focused on the possible mechanisms/hypothetical models of Z-DNA transition and its implications in AD. We propose that Z-DNA is formed in the promoter region of the APP, and Presenilin genes and Z-DNA may absorb the negative supercoils at that region. This decreases the supercoil density, altering the domain's native supercoiling state and facilitates the binding of effectors, which positively regulate gene expression of AD-related genes like APP and Presenilin. Further, it is presumed that Z-DNA may be involved in the down regulation of genes involved in Abeta clearance, anti-oxidant and defense mechanisms in AD. The proposed working model is novel and reveals possible triggering factors or precursors, which regulate the modulation of the supercoiling level of DNA involving putative Z-DNA forming sequences and regulatory proteins binding to DNA in AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , DNA, Z-Form/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Gene Expression Regulation , Humans , Models, Biological , Promoter Regions, Genetic/genetics , Transcription, Genetic
4.
Curr Pharm Des ; 10(25): 3111-9, 2004.
Article in English | MEDLINE | ID: mdl-15544501

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive dementia caused by the loss of the presynaptic markers of the cholinergic system in the brain areas related to memory and learning and brain deposits of amyloid beta peptide (A beta) and neurofibrillary tangles (NFT). A small fraction of early onset familial AD (FAD) is caused by mutations in genes, such as the beta-amyloid precursor protein (APP) and presenilins that increase the load of A beta in the brain. These studies together with findings that A beta is neurotoxic in vitro, provide evidence that some aggregates of this peptide are the key to the pathogenesis of AD. The yield of A beta and the processing and turnover of APP are regulated by a number of pathways including apolipoprotein E, cholesterol and cholinergic agonists. Early studies showed that muscarinic agonists increased APP processing within the A beta sequence (sAPP alpha). More recently, we have presented evidence showing that some, but not all, anticholinesterases reduce secretion of sAPP alpha as well as A beta into the media suggesting that cholinergic agonists modulate A beta levels by multiple mechanisms. Herein we review the recent advances in understanding the function of cholinesterase (ChE) in the brain and the use of ChE-inhibitors in AD. We propose and support the position that the influence of cholinergic stimulation on amyloid formation is critical in light of the early targeting of the cholinergic basal forebrain in AD and the possibility that maintenance of this cholinergic tone might slow amyloid deposition. In this context, the dual action of certain cholinesterase inhibitors on their ability to increase acetylcholine levels and decrease amyloid burden assumes significance as it may identify a single drug to both arrest the progression of the disease as well as treat its symptoms. A new generation of acetyl- and butyryl cholinesterase inhibitors is being studied and tested in human clinical trials for AD. We critically discuss recent trends in AD research, from molecular and genetic to clinical areas, as it relates to the effects of cholinergic agents and their secondary effects on A beta. Finally, we examine different neurobiological mechanisms that provide the basis of new targets for AD drug development.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Technology, Pharmaceutical/methods , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Drug Design , Humans , Technology, Pharmaceutical/trends
5.
Ann N Y Acad Sci ; 973: 334-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12485889

ABSTRACT

Two major pathological hallmarks of Alzheimer's disease (AD) are the senile plaques that are primarily composed of amyloid beta-peptide (Abeta) and neurofibrillary tangles consisting of tau aggregates. Abeta is generated proteolytically from a family of Abeta-containing precursor proteins (APP; 695-770 amino acid) by secretase enzymes to different specific carboxyl-terminal fragments (CTFs). Herein we examined APP and its products in autopsied brain sections from 10 AD and 10 non-AD control subjects immunochemically using an antibody that was raised against APP751-770 residue (O443). The O443 antibody was initially characterized by Western blot analysis and immunoprecipitation. In this study, we used this antibody for immunohistochemical analysis to determine the distribution of APP and its CTF species. In 10 brain regions showing different levels of plaques and tangles, antibody O443 stained the perinuclear region of the nucleus, plaques, and neurites. Tangle-bearing neurons also appeared to stain with the antibody, suggesting that these dysfunctional neurons continue to synthesize APP/CTF. Alternatively, the normally short-lived APP/CTF can be stabilized and persist in these neurons. Taken together, these results suggest that, in addition to the widely believed role of Abeta, CTFs may play a key role in the pathogenesis of AD. Studying their localization and biogenesis may reveal the biological activities of CTFs of APP. The present study may pave the way for possible antiamyloidogenic therapy in the treatment of AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/physiology , Brain/pathology , Peptide Fragments/physiology , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Animals , Antibody Specificity , Autopsy , Brain/metabolism , CHO Cells , Cricetinae , Humans , Immunohistochemistry , Molecular Sequence Data , Peptide Fragments/chemistry
7.
Proc Natl Acad Sci U S A ; 98(13): 7605-10, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11404470

ABSTRACT

The reduction in levels of the potentially toxic amyloid-beta peptide (Abeta) has emerged as one of the most important therapeutic goals in Alzheimer's disease. Key targets for this goal are factors that affect the expression and processing of the Abeta precursor protein (betaAPP). Earlier reports from our laboratory have shown that a novel cholinesterase inhibitor, phenserine, reduces betaAPP levels in vivo. Herein, we studied the mechanism of phenserine's actions to define the regulatory elements in betaAPP processing. Phenserine treatment resulted in decreased secretion of soluble betaAPP and Abeta into the conditioned media of human neuroblastoma cells without cellular toxicity. The regulation of betaAPP protein expression by phenserine was posttranscriptional as it suppressed betaAPP protein expression without altering betaAPP mRNA levels. However, phenserine's action was neither mediated through classical receptor signaling pathways, involving extracellular signal-regulated kinase or phosphatidylinositol 3-kinase activation, nor was it associated with the anticholinesterase activity of the drug. Furthermore, phenserine reduced expression of a chloramphenicol acetyltransferase reporter fused to the 5'-mRNA leader sequence of betaAPP without altering expression of a control chloramphenicol acetyltransferase reporter. These studies suggest that phenserine reduces Abeta levels by regulating betaAPP translation via the recently described iron regulatory element in the 5'-untranslated region of betaAPP mRNA, which has been shown previously to be up-regulated in the presence of interleukin-1. This study identifies an approach for the regulation of betaAPP expression that can result in a substantial reduction in the level of Abeta.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cholinesterase Inhibitors/pharmacology , Interleukin-1/pharmacology , Physostigmine/pharmacology , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , 5' Untranslated Regions/genetics , Astrocytoma , Cell Survival/drug effects , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/genetics , Chromones/pharmacology , Culture Media, Conditioned , Drug Design , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Humans , Interleukin-1/physiology , L-Lactate Dehydrogenase/analysis , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Neuroblastoma , Phosphatidylinositol 3-Kinases/metabolism , Physostigmine/analogs & derivatives , RNA, Messenger/metabolism , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
8.
Mol Pharmacol ; 59(3): 619-26, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179458

ABSTRACT

The beta-site amyloid precursor protein-cleaving enzyme (BACE) cleaves the amyloid precursor protein to produce the N terminus of the amyloid beta peptide, a major component of the plaques found in the brains of Alzheimer's disease patients. Sequence analysis of BACE indicates that the protein contains the consensus sequences found in most known aspartyl proteases, but otherwise has only modest homology with aspartyl proteases of known three-dimensional structure (i.e., pepsin, renin, or cathepsin D). Because BACE has been shown to be one of the two proteolytic activities responsible for the production of the Abeta peptide, this enzyme is a prime target for the design of therapeutic agents aimed at reducing Abeta for the treatment of Alzheimer's disease. Toward this ultimate goal, we have expressed a recombinant, truncated human BACE in a Drosophila melanogaster S2 cell expression system to generate high levels of secreted BACE protein. The protein was convenient to purify and was enzymatically active and specific for cleaving the beta-secretase site of human APP, as demonstrated with soluble APP as the substrate in novel sandwich enzyme-linked immunosorbent assay and Western blot assays. Further kinetic analysis revealed no catalytic differences between this recombinant, secreted BACE, and brain BACE. Both showed a strong preference for substrates that contained the Swedish mutation, where NL is substituted for KM immediately upstream of the cleavage site, relative to the wild-type sequence, and both showed the same extent of inhibition by a peptide-based inhibitor. The capability to produce large quantities of BACE enzyme will facilitate protein structure determination and inhibitor development efforts that may lead to the evolution of useful Alzheimer's disease treatments.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Peptide Hydrolases/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Chromatography, High Pressure Liquid , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Endopeptidases , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Solubility , Transfection
9.
J Biol Chem ; 276(1): 481-7, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11035007

ABSTRACT

Alzheimer's disease is characterized by the deposits of the 4-kDa amyloid beta peptide (A beta). The A beta protein precursor (APP) is cleaved by beta-secretase to generate a C-terminal fragment, CTF beta, which in turn is cleaved by gamma-secretase to generate A beta. Alternative cleavage of the APP by alpha-secretase at A beta 16/17 generates the C-terminal fragment, CTFalpha. In addition to A beta, endoproteolytic cleavage of CTF alpha and CTF beta by gamma-secretase should yield a C-terminal fragment of 57-59 residues (CTF gamma). However, CTF gamma has not yet been reported in either brain or cell lysates, presumably due to its instability in vivo. We detected the in vitro generation of A beta as well as an approximately 6-kDa fragment from guinea pig brain membranes. We have provided biochemical and pharmacological evidence that this 6-kDa fragment is the elusive CTF gamma, and we describe an in vitro assay for gamma-secretase activity. The fragment migrates with a synthetic peptide corresponding to the 57-residue CTF gamma fragment. Three compounds previously identified as gamma-secretase inhibitors, pepstatin-A, MG132, and a substrate-based difluoroketone (t-butoxycarbonyl-Val-Ile-(S)-4-amino-3-oxo-2, 2-difluoropentanoyl-Val-Ile-OMe), reduced the yield of CTF gamma, providing additional evidence that the fragment arises from gamma-secretase cleavage. Consistent with reports that presenilins are the elusive gamma-secretases, subcellular fractionation studies showed that presenilin-1, CTF alpha, and CTF beta are enriched in the CTF gamma-generating fractions. The in vitro gamma-secretase assay described here will be useful for the detailed characterization of the enzyme and to screen for gamma-secretase inhibitors.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Peptide Fragments/analysis , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/chemistry , Animals , Brain/cytology , Brain/enzymology , Brain/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Detergents/pharmacology , Endopeptidases/analysis , Guinea Pigs , Hydrogen-Ion Concentration , Membrane Proteins/analysis , Membrane Proteins/metabolism , Molecular Weight , Pepstatins/pharmacology , Peptide Fragments/chemistry , Phenanthrolines/pharmacology , Protease Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Solubility/drug effects , Subcellular Fractions/metabolism
10.
FASEB J ; 15(1): 16-18, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11099491

ABSTRACT

The abnormal accumulation of the amyloid beta protein (Abeta) has been implicated as an early and critical event in the etiology and pathogenesis of Alzheimer's disease (AD). Compounds that reduce Abeta accumulation may therefore be useful therapeutically. In cell-based screens we detected a significant reduction in Abeta concentration after treatment with the phosphatidylinositol kinase inhibitors wortmannin and LY294002. To determine the effect of this class of compounds on in vivo Abeta accumulation, we administered wortmannin to the Tg2576 mouse model of AD. Oral administration of wortmannin over four months resulted in a significant, non-overlapping 40%-50% reduction in the number of senile plaques, one of the pathological hallmarks of AD. Sandwich ELISA analysis of formic acid extractable Abeta in the brain of treated animals indicates that both Abeta40 and the longer, more amyloidogenic form of the peptide, Abeta42, were significantly reduced. These data provide the first direct evidence that compounds identified by their ability to reduce Abeta concentration in vitro can reduce Abeta accumulation and deposition in the brain, thus establishing a basic paradigm for the identification and evaluation of additional compounds that lower Abeta accumulation.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Androstadienes/administration & dosage , Androstadienes/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Administration, Oral , Aging/physiology , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Androstadienes/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Mice , Mice, Transgenic , Models, Biological , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plaque, Amyloid/drug effects , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Solubility , Wortmannin
11.
FASEB J ; 14(15): 2383-6, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11024004

ABSTRACT

The amyloid b-protein (Ab) deposited in Alzheimer's disease (AD) is a normally secreted proteolytic product of the amyloid b-protein precursor (APP). Generation of Ab from the APP requires two sequential proteolytic events: an initial b-secretase cleavage at the amino terminus of the Ab sequence followed by g-secretase cleavage at the carboxyl terminus of Ab. We describe the development of a robust in vitro assay for g-secretase cleavage by showing de novo Ab production in vitro and establish that this assay monitors authentic gamma-secretase activity by documenting the production of a cognate g-CTF, confirming the size of the Ab produced by mass spectrometry, and inhibiting cleavage in this system with multiple inhibitors that alter g-secretase activity in living cells. Using this assay, we demonstrate that the g-secretase activity 1) is tightly associated with the membrane, 2) can be solubilized, 3) has a pH optimum of 6.8 but is active from pH 6.0 to pH >8.4, and 4) ascertain that activities of the g-40 and g-42 are indeed pharmacologically distinct. These studies should facilitate the purification of the protease or proteases that are responsible for this unusual activity, which is a major therapeutic target for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/analysis , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Animals , CHO Cells , Cell-Free System , Cricetinae , Hydrogen-Ion Concentration , Models, Biological , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Solubility
12.
Neurobiol Dis ; 7(4): 321-31, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10964604

ABSTRACT

Recent data suggest that cholesterol metabolism is linked to susceptibility to Alzheimer's disease (AD). However, no direct evidence has been reported linking cholesterol metabolism and the pathogenesis of AD. To test the hypothesis that amyloid beta-peptide (Abeta) deposition can be modulated by diet-induced hypercholesterolemia, we used a transgenic-mouse model for AD amyloidosis and examined the effects of a high-fat/high-cholesterol diet on central nervous system (CNS) Abeta accumulation. Our data showed that diet-induced hypercholesterolemia resulted in significantly increased levels of formic acid-extractable Abeta peptides in the CNS. Furthermore, the levels of total Abeta were strongly correlated with the levels of both plasma and CNS total cholesterol. Biochemical analysis revealed that, compared with control, the hypercholesterolemic mice had significantly decreased levels of sAPPalpha and increased levels of C-terminal fragments (beta-CTFs), suggesting alterations in amyloid precursor protein processing in response to hypercholesterolemia. Neuropathological analysis indicated that the hypercholesterolemic diet significantly increased beta-amyloid load by increasing both deposit number and size. These data demonstrate that high dietary cholesterol increases Abeta accumulation and accelerates the AD-related pathology observed in this animal model. Thus, we propose that diet can be used to modulate the risk of developing AD.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Cholesterol, Dietary/adverse effects , Disease Models, Animal , Hypercholesterolemia/complications , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/blood , Hypercholesterolemia/blood , Hypercholesterolemia/etiology , Mice , Mice, Transgenic
13.
J Neurochem ; 73(6): 2383-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10582597

ABSTRACT

Autosomal dominant mutations in the presenilin 1 (PS1) gene are associated with familial, early-onset Alzheimer's disease. Although the pathogenic mechanism of these mutations is unclear, their common feature is that they lead to an increased concentration of amyloid beta-peptide (Abeta) 42 in the plasma of early-onset patients, in the conditioned media of transfected cells, and in the brains of transgenic mice that overexpress mutant PS1. To address the mechanism(s) by which the pathogenic PS1 mutations increase Abeta42, we constructed human cell lines expressing a doxycyclin (dox)-inducible antisense PS1 RNA and measured its effects on the levels of PS1, amyloid precursor protein (APP), and Abeta. In time course experiments, we observed a statistically significant (p = 0.0038) more than twofold elevation in secreted Abeta42 as early as 12 days after addition of dox. This correlated with an 80% decrease in the 46-kDa PS1 holoprotein and a 30% decrease in the 26-kDa N-terminal fragment (NTF). Furthermore, there was a significant fivefold (p = 0.002) increase in Abeta42 after 14-day dox treatment; this correlated with a >90% decrease in PS1 holoprotein and 60% decrease in NTF. At no time point did we observe significant changes in Abeta40, APP holoprotein, presenilin 2, or tubulin. Ten days after the removal of dox, we observed a return to constitutive levels for Abeta42, PS1 holoprotein, and NTF. These results suggest that in human cell lines, the reduction of normal PS1 activity results in the increased production of Abeta42. Furthermore, our results are consistent with a loss of function or dominant negative mechanism for the pathogenic PS1 mutations.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/biosynthesis , Gene Expression Regulation/drug effects , Membrane Proteins/metabolism , Peptide Fragments/biosynthesis , RNA, Antisense/pharmacology , Amyloid beta-Peptides/genetics , Cell Line , Doxycycline/pharmacology , Humans , Kidney , Membrane Proteins/genetics , Peptide Fragments/genetics , Presenilin-1 , Transfection , Tubulin/biosynthesis , Tubulin/genetics
14.
Biochem J ; 343 Pt 3: 627-35, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10527942

ABSTRACT

Glycosylphosphatidylinositol (GPI)-anchored proteins are resistant to solubilization with Triton X-100 at 4 degrees C, and they can be recovered in Triton-insoluble membranes (TIMs) that float to a characteristic buoyant density. Because the GPI structure itself has been shown to target GPI-anchored proteins to TIMs, we investigated the association of GPI-anchor intermediates with TIMs. GPI-anchor biosynthesis involves a pathway of some 10 steps that take place in the endoplasmic reticulum (ER). These intermediates include glucosaminyl-acylphosphatidylinositol [GlcN-(acyl)PI] and later mannosylated GPIs, denoted H6, H7 and H8, that are present not only in the ER but also in other cell compartments, including the plasma membrane. At least two-thirds of the GlcN-(acyl)PI in HeLa D cells and mannosylated GPIs in K562 cells were found in TIMs. Although previous reports have considered TIMs to be derived primarily from the plasma membrane, we recovered TIMs from subcellular fractions enriched in ER membranes. The ER marker calnexin and GPI-anchored proteins as well as N-acetylglucosaminyl-phosphatidylinositol and mannosylated GPIs were present in ER-TIMs. Interestingly, GlcN-PI and H7 were less enriched in ER-TIM than the other GPIs, suggesting that ER-TIMs might reflect a compartmentalization of the GPI-anchor biosynthetic pathway in the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Glycosylphosphatidylinositols/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Animals , CHO Cells , Carbohydrate Sequence , Cell Fractionation , Choline/metabolism , Cricetinae , Cysteine/metabolism , Endoplasmic Reticulum/ultrastructure , Glucosamine/metabolism , HeLa Cells , Humans , Inositol/metabolism , Intracellular Membranes/ultrastructure , K562 Cells , Mannose/metabolism , Membrane Proteins/isolation & purification , Models, Chemical , Molecular Sequence Data , Polyethylene Glycols , Radioisotope Dilution Technique , Sulfur Radioisotopes , Tritium , Uridine Diphosphate N-Acetylglucosamine/metabolism
15.
J Biol Chem ; 274(38): 26810-4, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480887

ABSTRACT

The Alzheimer's amyloid protein (Abeta) is released from the larger amyloid beta-protein precursor (APP) by unidentified enzymes referred to as beta- and gamma-secretase. beta-Secretase cleaves APP on the amino side of Abeta producing a large secreted derivative (sAPPbeta) and an Abeta-bearing C-terminal derivative that is subsequently cleaved by gamma-secretase to release Abeta. Alternative cleavage of the APP by alpha-secretase at Abeta16/17 releases the secreted derivative sAPPalpha. In yeast, alpha-secretase activity has been attributed to glycosylphosphatidylinositol (GPI)-anchored aspartyl proteases. To examine the role of GPI-anchored proteins, we specifically removed these proteins from the surface of mammalian cells using phosphatidylinositol-specific phospholipase C (PI-PLC). PI-PLC treatment of fetal guinea pig brain cultures substantially reduced the amount of Abeta40 and Abeta42 in the medium but had no effect on sAPPalpha. A mutant CHO cell line (gpi85), which lacks GPI-anchored proteins, secreted lower levels of Abeta40, Abeta42, and sAPPbeta than its parental line (GPI+). When this parental line was treated with PI-PLC, Abeta40, Abeta42, and sAPPbeta decreased to levels similar to those observed in the mutant line, and the mutant line was resistant to these effects of PI-PLC. These findings provide strong evidence that one or more GPI-anchored proteins play an important role in beta-secretase activity and Abeta secretion in mammalian cells. The cell-surface GPI-anchored protein(s) involved in Abeta biogenesis may be excellent therapeutic target(s) in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Glycosylphosphatidylinositols/metabolism , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Brain/enzymology , CHO Cells , Cells, Cultured , Cricetinae , Endopeptidases/metabolism , Enzyme Activation , Guinea Pigs , Humans , Hydrolysis , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Protein Kinase C/metabolism , Type C Phospholipases/metabolism
16.
Brain Res Mol Brain Res ; 62(2): 131-40, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9813282

ABSTRACT

The amyloid beta-protein (Abeta) is an approximately 4 kD secreted protein normally found in human plasma and cerebrospinal fluid. Abeta is invariably deposited as insoluble amyloid fibrils in the brains of patients with Alzheimer's disease (AD), and there is increasing evidence that Abeta deposition plays an important role in AD pathogenesis. Abeta is released from the larger beta-amyloid precursor protein (betaAPP) through cleavage on the amino and carboxyl side of Abeta by proteolytic activities referred to as beta and gamma secretase, respectively. betaAPP is also cleaved at Abeta16 by a third protease, alpha secretase, which may prevent amyloid deposition by bisecting the Abeta peptide. Tacrine, a cholinesterase inhibitor, has been shown to improve memory and cognitive functions in some patients with AD, and we have previously demonstrated that it significantly reduces the levels of the secretion of soluble betaAPP fragments (sAPP) in cultured cells. In this study, we extended our studies by analysis of Abeta40 and Abeta42 and report that in a human neuroblastoma cell line tacrine reduced the levels of total Abeta, Abeta40 and Abeta42 in addition to sAPP. These inhibitory results cannot be attributed to a reduction in total betaAPP synthesis as tacrine treatment did not cause a significant change in the rate of betaAPP synthesis. Furthermore, significant toxicity was not observed in tacrine-treated cultures as determined by analysis of lactate dehydrogenase (LDH) in the conditioned media. Taken together, these results suggest that tacrine affects the processing of betaAPP by alterations in betaAPP trafficking and/or increased intracellular proteolysis. This study raises the possibility that tacrine may aid in the treatment of AD due to its effects on betaAPP processing as well as by its effects on the cholinergic pathway.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/drug effects , Peptide Fragments/metabolism , Tacrine/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Biological Transport/drug effects , Culture Media, Conditioned , Endopeptidases/metabolism , Humans , L-Lactate Dehydrogenase/analysis , Neuroblastoma/pathology , Neurons/metabolism , Protein Processing, Post-Translational/drug effects , Solubility , Tumor Cells, Cultured
17.
Biochem Biophys Res Commun ; 249(2): 546-9, 1998 Aug 19.
Article in English | MEDLINE | ID: mdl-9712734

ABSTRACT

The 42-residue amyloid beta protein (Abeta42) has been shown to be toxic to neurons and is believed to play a key causative role in Alzheimer's disease (AD). A search for Abeta binding proteins that can mediate its toxicity resulted in the identification of the endoplasmic-reticulum (ER) associated Abeta binding protein (ERAB) which was also shown to be involved in Abeta induced apoptosis. The primary report indicated that a signal sequence is absent in ERAB suggesting that it is bound to the cytoplasmic aspect of cellular membranes. Abeta is generated in the lumen of secretory organelles and released into the medium resulting in its separation from ERAB by a membrane barrier. After computer analysis of the ERAB sequence, we have detected a putative signal peptide that can direct the protein into the secretory pathway. This signal sequence was found in human, rodent, and bovine ERAB suggesting that it is a type II integral membrane protein in vertebrates. This topology can explain the binding of Abeta to ERAB. Our finding that an integral membrane form of ERAB can bind to Abeta in the lumen of transport vesicles and other cytoplasmic receptors provides a basis for understanding its role in AD.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases , Carrier Proteins/chemistry , Protein Sorting Signals/analysis , Alzheimer Disease , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Animals , Cattle , Drosophila , Humans , Membrane Proteins/chemistry , Mice , Microbodies/chemistry , Mitochondria/chemistry , Software
18.
Biochemistry ; 37(5): 1274-82, 1998 Feb 03.
Article in English | MEDLINE | ID: mdl-9477953

ABSTRACT

We previously demonstrated the presence of a soluble form of full-length Alzheimer's amyloid precursor protein (APP) in the lumen of adrenal medullary chromaffin granules (CG). Furthermore, full-length APP is released from CG membranes in vitro at pH 9.0 by an enzymatic mechanism, sensitive to protease inhibitors [Vassilacopoulou et al. (1995) J. Neurochem. 64, 2140-2146]. In this study, we found that when intact CG were subjected to exogenous trypsin, a fraction of APP was not digested, consistent with an intragranular population of APP. To examine the substrate-product relationship between membrane and soluble full-length APP, we labeled CG transmembrane APP with 3-(trifluoromethyl)-3-(m-[125I]iodophenyl)diazirine ([125I]TID), a lipophilic probe, specific for membrane-spanning domains of proteins. APP released from the membranes at pH 9.0 was not labeled with [125I]TID. In addition, this APP was not biotinylated in intact CG. Combined, the results indicate that APP released from CG membranes derives from a unique nontransmembrane population of membrane-associated APP, located in the lumenal side of CG membranes. Dithiobis(succinimidylpropionate) (DSP) cross-linking indicated that APP in CG is situated in close proximity with other proteins, possibly with APP itself. APP complexes were also detected under nonreducing conditions, without DSP cross-linking. These results, combined with our previous studies, indicate that full-length APP within CG exists as three different populations: (I) transmembrane, (II) membrane-associated/nontransmembrane, and (III) soluble. The existence of nontransmembrane populations suggests that putative gamma-secretase cleavage sites of APP, assumed to be buried within the lipid bilayer, could be accessible to proteolysis in a soluble intravesicular environment.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Chromaffin Granules/metabolism , Membrane Proteins/metabolism , Adrenal Medulla , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/drug effects , Animals , Azirines/metabolism , Biotinylation , Cattle , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Chromaffin Granules/chemistry , Chromaffin Granules/drug effects , Cross-Linking Reagents , Humans , Iodine Radioisotopes , Membrane Proteins/chemistry , Photoaffinity Labels , Trypsin/pharmacology
19.
Life Sci ; 61(20): 1985-92, 1997.
Article in English | MEDLINE | ID: mdl-9366505

ABSTRACT

The senile plaque in Alzheimer's disease (AD) consists mainly of the amyloid beta-peptide (A beta) derived from a larger beta-amyloid precursor protein (betaAPP). The majority of betaAPP is processed by either a secretory or lysosomal/endosomal pathway. Soluble derivatives of betaAPP (sAPP) and A beta generated by the proteolytic processing of full-length betaAPP are normally secreted into the conditioned medium of cultured cells. Tacrine, a centrally active potent cholinesterase inhibitor that has been shown to improve cognitive functions in some patients with AD, inhibits the secretion of sAPP. Here we have investigated whether leupeptin, a lysosomal protease inhibitor, could influence this effect of tacrine. We analyzed levels of betaAPP derivatives in cultured HeLa cells by immunoblotting cell lysates and conditioned media using the monoclonal antibody 22C11. Levels of sAPP normally present in conditioned media were severely reduced by treating cells with tacrine. The treatment of cells with tacrine resulted in a small decrease in the intracellular levels of betaAPP. The effect of treating the cells with tacrine did not depend upon the growing state of the cells as a similar effect was observed when the drug was added either during initial plating of the cells or after the attachment of the cells. The effect of tacrine was not affected by preincubating the cells with low serum in the culture medium. The treatment of cells with tacrine plus leupeptin reduced the secretion of sAPP in the medium to the same degree as did the treatment with tacrine alone, suggesting that the tacrine-mediated inhibition of sAPP release may not involve leupeptin-sensitive proteolytic pathways. The results suggest that the inhibitory effect of tacrine on sAPP secretion is not due to the proteolytic cleavage of the holoprotein in the medium.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cholinesterase Inhibitors/pharmacology , HeLa Cells/drug effects , HeLa Cells/metabolism , Leupeptins/pharmacology , Tacrine/pharmacology , Alzheimer Disease , Carbachol/pharmacology , Culture Media, Conditioned , Humans , Immunoblotting , Parasympathomimetics/pharmacology , Protease Inhibitors/pharmacology
20.
Mol Chem Neuropathol ; 28(1-3): 21-34, 1996.
Article in English | MEDLINE | ID: mdl-8871938

ABSTRACT

Involvement of free-radical oxidations in the aging process has been a topic of interest since Harman's original contribution. Because of the close association between aging and Alzheimer disease (AD) and the qualitative similarity in the neuropathology of both conditions, it has been proposed by many investigators that oxidative stress may be important in Ad. If such modality of injury was indeed involved, one should expect to find markers of oxidation and heat shock (since free radicals are key mediators of heat-shock induction) in brains of patients with AD. In fact, several studies documented abnormal expression of antioxidant enzymes and heat-shock proteins (HSP) along with other markers of oxidation in AD brains. We showed that abnormally expressed antioxidant enzymes are topographically associated with senile plaques and neurofibrillary tangles, and that the activity of these enzymes is (contrary to what one would expect) markedly reduced. These findings have recently been confirmed by other investigators. Despite a large amount of evidence that suggests an association between oxidative stress and the pathogenesis of AD, it is not yet known whether oxidative stress is a cause or consequence of the disorder. Future research efforts regarding the oxidative stress hypothesis of AD should include attempts at generating AD pathology by oxidative means in laboratory animals, determining the role and integrity of the heat-shock response in AD, as well as that of various antioxidant systems, growth factors, and hormones with antioxidant and neuroprotective properties.


Subject(s)
Alzheimer Disease/physiopathology , Brain/metabolism , Heat-Shock Proteins/biosynthesis , Oxidative Stress , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Animals , Antioxidants/metabolism , Brain/pathology , Hot Temperature , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...