Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Mol Neurodegener ; 14(1): 45, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31791377

ABSTRACT

Organotypic slice cultures of brain or spinal cord have been a longstanding tool in neuroscience research but their utility for understanding Alzheimer's disease (AD) and other neurodegenerative proteinopathies has only recently begun to be evaluated. Organotypic brain slice cultures (BSCs) represent a physiologically relevant three-dimensional model of the brain. BSCs support all the central nervous system (CNS) cell types and can be produced from brain areas involved in neurodegenerative disease. BSCs can be used to better understand the induction and significance of proteinopathies underlying the development and progression of AD and other neurodegenerative disorders, and in the future may serve as bridging technologies between cell culture and in vivo experiments for the development and evaluation of novel therapeutic targets and strategies. We review the initial development and general use of BSCs in neuroscience research and highlight the advantages of these cultures as an ex vivo model. Subsequently we focus on i) BSC-based modeling of AD and other neurodegenerative proteinopathies ii) use of BSCs to understand mechanisms underlying these diseases and iii) how BSCs can serve as tools to screen for suitable therapeutics prior to in vivo investigations. Finally, we will examine i) open questions regarding the use of such cultures and ii) how emerging technologies such as recombinant adeno-associated viruses (rAAV) may be combined with these models to advance translational research relevant to neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Animals , Disease Models, Animal , Humans , Organ Culture Techniques/methods
2.
Neuroimage Clin ; 22: 101800, 2019.
Article in English | MEDLINE | ID: mdl-30991618

ABSTRACT

The threshold for amyloid positivity by visual assessment on PET has been validated by comparison to amyloid load measured histopathologically and biochemically at post mortem. As such, it is now feasible to use qualitative visual assessment of amyloid positivity as an in-vivo gold standard to determine those factors which can modify the quantitative threshold for amyloid positivity. We calculated quantitative amyloid load, measured as Standardized Uptake Value Ratios (SUVRs) using [18-F]florbetaben PET scans, for 159 Hispanic and non-Hispanic participants, who had been classified clinically as Cognitively Normal (CN), Mild Cognitive Impairment (MCI) or Dementia (DEM). PET scans were visually rated as amyloid positive (A+) or negative (A-), and these judgments were used as the gold standard with which to determine (using ROC analyses) the SUVR threshold for amyloid positivity considering factors such as age, ethnicity (Hispanic versus non-Hispanic), gender, cognitive status, and apolipoprotein E ε4 carrier status. Visually rated scans were A+ for 11% of CN, 39.0% of MCI and 70% of DEM participants. The optimal SUVR threshold for A+ among all participants was 1.42 (sensitivity = 94%; specificity = 92.5%), but this quantitative threshold was higher among E4 carriers (SUVR = 1.52) than non-carriers (SUVR = 1.31). While mean SUVRs did not differ between Hispanic and non-Hispanic participants;, a statistically significant interaction term indicated that the effect of E4 carrier status on amyloid load was greater among non-Hispanics than Hispanics. Visual assessment, as the gold standard for A+, facilitates determination of the effects of various factors on quantitative thresholds for amyloid positivity. A continuous relationship was found between amyloid load and global cognitive scores, suggesting that any calculated threshold for the whole group, or a subgroup, is artefactual and that the lowest calculated threshold may be optimal for the purposes of early diagnosis and intervention.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Cognitive Dysfunction , Dementia , Hispanic or Latino , Neuroimaging/standards , Age Factors , Aged , Aged, 80 and over , Aniline Compounds , Cognitive Dysfunction/ethnology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Dementia/ethnology , Dementia/genetics , Dementia/metabolism , Dementia/physiopathology , Female , Hispanic or Latino/genetics , Hispanic or Latino/statistics & numerical data , Humans , Male , Middle Aged , Positron-Emission Tomography/standards , Sensitivity and Specificity , Sex Factors , Stilbenes
3.
Oncogene ; 27(38): 5124-31, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18758481

ABSTRACT

Accumulating preclinical and clinical evidence supports a pro-oncogenic function for Notch signaling in several solid tumors, particularly but not exclusively in breast cancer. Notch inhibitory agents, such as gamma-secretase inhibitors, are being investigated as candidate cancer therapeutic agents. Interest in therapeutic modulation of the Notch pathway has been increased by recent reports, indicating that its role is important in controlling the fate of putative 'breast cancer stem cells'. However, as is the case for most targeted therapies, successful targeting of Notch signaling in cancer will require a considerable refinement of our understanding of the regulation of this pathway and its effects in both normal and cancer cells. Notch signaling has bidirectional 'cross talk' interaction with multiple other pathways that include candidate therapeutic targets. Understanding these interactions will greatly increase our ability to design rational combination regimens. To determine which patients are most likely to benefit from treatment with Notch inhibitors, it will be necessary to develop molecular tests to accurately measure pathway activity in specific tumors. Finally, mechanism-based toxicities will have to be addressed by a careful choice of therapeutic agents, combinations and regimens. This article summarizes the current state of the field, and briefly describes opportunities and challenges for Notch-targeted therapies in oncology.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Notch/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Division/drug effects , Cell Division/physiology , Combined Modality Therapy , Drug Delivery Systems , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms/physiopathology , Neoplasms/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Tumor Suppressor Proteins/physiology
4.
Oncogene ; 27(37): 5019-32, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18469855

ABSTRACT

ErbB-2 overexpression in breast tumors is associated with poor survival. Expression of Notch-1 and its ligand, Jagged-1, is associated with the poorest survival, including ErbB-2-positive tumors. Trastuzumab plus chemotherapy is the standard of care for ErbB-2-positive breast cancer. A proportion of tumors are initially resistant to trastuzumab and acquired resistance to trastuzumab occurs in metastatic breast cancer and is associated with poor prognosis. Thus, we investigated whether Notch-1 contributes to trastuzumab resistance. ErbB-2-positive cells have low Notch transcriptional activity compared to non-overexpressing cells. Trastuzumab or a dual epidermal growth factor receptor (EGFR)/ErbB-2 tyrosine kinase inhibitor (TKI) increased Notch activity by 2- to 6-fold in SKBr3, BT474 and MCF-7/HER2-18 cells. The increase in activity was abrogated by a Notch inhibitor, gamma-secretase inhibitor (GSI) or Notch-1 small-interfering RNA (siRNA). Trastuzumab decreased Notch-1trade mark precursor, increased amount and nuclear accumulation of active Notch-1(IC) and increased expression of targets, Hey1 and Deltex1 mRNAs, and Hes5, Hey1, Hes1 proteins. Importantly, trastuzumab-resistant BT474 cells treated with trastuzumab for 6 months expressed twofold higher Notch-1, twofold higher Hey1, ninefold higher Deltex1 mRNAs and threefold higher Notch-1 and Hes5 proteins, compared to trastuzumab-sensitive BT474 cells. The increase in Hey1 and Deltex1 mRNAs in resistant cells was abrogated by a Notch-1 siRNA. Cell proliferation was inhibited more effectively by trastuzumab or TKI plus a GSI than either agent alone. Decreased Notch-1 by siRNA increased efficacy of trastuzumab in BT474 sensitive cells and restored sensitivity in resistant cells. Trastuzumab plus a GSI increased apoptosis in sensitive cells by 20-30%. A GSI alone was sufficient to increase apoptosis in trastuzumab-resistant BT474 cells by 20%, which increased to 30% with trastuzumab. Notch-1 siRNA alone decreased cell growth by 30% in sensitive and more than 50% in resistant BT474 cells. Furthermore, growth of both trastuzumab sensitive and resistant cells was completely inhibited by combining trastuzumab plus Notch-1 siRNA. More importantly, Notch-1 siRNA or a GSI resensitized trastuzumab-resistant BT474 cells to trastuzumab. These results demonstrate that ErbB-2 overexpression suppresses Notch-1 activity, which can be reversed by trastuzumab or TKI. These results suggest that Notch-1 might play a novel role in resistance to trastuzumab, which could be prevented or reversed by inhibiting Notch-1.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, Notch1/metabolism , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , RNA, Small Interfering/pharmacology , Receptor, ErbB-2/immunology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/physiology , Trastuzumab , Tumor Cells, Cultured , Up-Regulation/drug effects
5.
J Neurochem ; 87(4): 1052-5, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14622134

ABSTRACT

As a normal consequence of aging, men experience a significant decline in androgen levels. Although the neural consequences of age-related androgen depletion remain unclear, recent evidence suggests a link between low androgen levels and the development of Alzheimer's disease (AD). Here, we test the hypothesis that androgens act as endogenous modulators of beta-amyloid protein (Abeta) levels. To investigate this possibility, brain and plasma levels of Abeta were measured in male rats with varying hormonal conditions. Depletion of endogenous sex steroid hormones via gonadectomy (GDX) resulted in increased brain levels of Abeta in comparison to gonadally intact male rats. This GDX-induced increase in Abeta levels was reversed by DHT supplementation, demonstrating a functional role for androgens in modulating brain levels of Abeta. These findings suggest that age-related androgen depletion may result in accumulation of Abeta in the male brain and thereby act as a risk factor for the development of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Androgens/physiology , Brain/drug effects , Brain/metabolism , Amyloid beta-Peptides/blood , Androgens/administration & dosage , Animals , Dihydrotestosterone/administration & dosage , Drug Implants , Estradiol/administration & dosage , Hormone Replacement Therapy , Male , Orchiectomy , Rats , Rats, Sprague-Dawley , Receptors, Androgen/metabolism , Septal Nuclei/cytology , Septal Nuclei/drug effects , Septal Nuclei/metabolism
6.
Dement Geriatr Cogn Disord ; 14(1): 13-21, 2002.
Article in English | MEDLINE | ID: mdl-12053127

ABSTRACT

We report a kindred with three cases of dementia. The proband presented with forgetfulness and personality changes at age 56, followed shortly thereafter by behavioral dyscontrol, hyperphagia, hypersexuality, delusions, illusions, disinhibition and double incontinence. Neuroimaging studies were consistent with frontotemporal dementia (FTD). In one allele, an arginine insertion at codon 352 in the presenilin 1 (PSEN1) gene was identified; no mutation was identified in the amyloid precursor protein or tau genes. We conclude that the clinical features of the Kluver-Bucy syndrome and FTD can be associated with PSEN1 mutations. Furthermore, presenilin analyses may be helpful to characterize kindreds with familial dementing illnesses regardless of the phenotype, particularly if no tau mutation is present.


Subject(s)
Dementia/genetics , Membrane Proteins/genetics , Mutation , Dementia/diagnosis , Dementia/psychology , Humans , Intelligence , Magnetic Resonance Imaging , Middle Aged , Neuropsychological Tests , Pedigree , Presenilin-1
7.
Brain Res Mol Brain Res ; 97(1): 103-13, 2001 Dec 16.
Article in English | MEDLINE | ID: mdl-11744168

ABSTRACT

The amyloid precursor protein (APP) is cleaved by two enzymes, beta-secretase and gamma-secretase, to generate the pathological amyloid beta (Abeta) peptide. Expression of familial Alzheimer's disease (FAD) mutants of APP in primary neurons causes both intracellular accumulation of the C-terminal beta-secretase cleavage product of APP and increased secretion of Abeta, and eventually results in apoptotic death of the cells. To determine whether either of these two processing products of APP is involved in this apoptotic pathway, we first modeled experimentally the accumulation of the beta-secretase cleavage product in neurons. The C-terminal 100 amino acids (C100) of APP, with and without a signal peptide, was expressed in cells via recombinant herpes simplex virus (HSV) vectors. Both transgene products were targeted to the membrane, and both caused apoptosis in the neurons, implicating the beta-secretase cleavage product of APP in apoptosis caused by FAD APPs. Expression in neurons of a mutant of FAD APP that inhibited beta-secretase cleavage inhibited its ability to cause apoptosis. However, expression in neurons of a mutant of FAD APP that inhibited gamma-secretase cleavage did not inhibit the ability of this mutant to cause apoptosis. These data suggested that the C-terminal beta-secretase cleavage product of APP, but not Abeta, mediates the apoptosis caused by FAD mutants of APP. Consistent with this hypothesis, C31, which is generated from the beta-secretase cleavage product, itself caused neuronal apoptosis. Inhibitors of caspases 3, 6 and 8, but not of caspase 9, inhibited the apoptosis caused by FAD mutants of APP. It may be inferred from these data that beta-secretase cleavage of FAD mutants of APP allows the appropriate caspase access to its site of action to produce C31, which directly causes neuronal apoptosis.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apoptosis/physiology , Aspartic Acid Endopeptidases/physiology , Nerve Tissue Proteins/physiology , Alzheimer Disease/genetics , Amino Acid Substitution , Amyloid Precursor Protein Secretases , Animals , Caspases/metabolism , Culture Media, Conditioned , Endopeptidases/metabolism , Genetic Vectors/genetics , Humans , London , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Peptide Fragments/metabolism , Point Mutation , Rats , Simplexvirus/genetics , Sweden , Transgenes
8.
Neurobiol Aging ; 22(5): 721-7, 2001.
Article in English | MEDLINE | ID: mdl-11705631

ABSTRACT

Vaccinations with Abeta1-42 have been shown to reduce amyloid burden in transgenic models of Alzheimer's disease (AD). We have further tested the efficacy of Abeta1-42 immunization in the Tg2576 mouse model of AD by immunizing one group of mice with minimal Abeta deposition, one group of mice with modest Abeta deposition, and one group with significant Abeta deposition. The effects of immunization on Abeta deposition were examined using biochemical and immunohistochemical methods. In Tg2576 mice immunized prior to significant amyloid deposition, Abeta1-42 immunization was highly effective. Biochemically extracted Abeta40 and Abeta42 levels were significantly reduced and immunohistochemical plaque load was also reduced. Immunization of mice with modest amounts of pre-existing Abeta deposits selectively reduced Abeta42 without altering Abeta40, although plaque load was reduced. In contrast, in Tg2576 mice with significant pre-existing Abeta loads, Abeta1-42 immunization only minimally decreased Abeta42 levels, whereas no alteration in Abeta40 levels or in plaque load was observed. These results indicate that in Tg2576 mice, Abeta1-42 immunization is more effective at preventing additional Abeta accumulation and does not result in significant clearance of pre-existing Abeta deposits.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/prevention & control , Peptide Fragments/immunology , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloidosis/pathology , Animals , Brain/pathology , Disease Models, Animal , Humans , Immunization , Mice , Mice, Transgenic , Plaque, Amyloid/pathology
9.
Nature ; 414(6860): 212-6, 2001 Nov 08.
Article in English | MEDLINE | ID: mdl-11700559

ABSTRACT

Epidemiological studies have documented a reduced prevalence of Alzheimer's disease among users of nonsteroidal anti-inflammatory drugs (NSAIDs). It has been proposed that NSAIDs exert their beneficial effects in part by reducing neurotoxic inflammatory responses in the brain, although this mechanism has not been proved. Here we report that the NSAIDs ibuprofen, indomethacin and sulindac sulphide preferentially decrease the highly amyloidogenic Abeta42 peptide (the 42-residue isoform of the amyloid-beta peptide) produced from a variety of cultured cells by as much as 80%. This effect was not seen in all NSAIDs and seems not to be mediated by inhibition of cyclooxygenase (COX) activity, the principal pharmacological target of NSAIDs. Furthermore, short-term administration of ibuprofen to mice that produce mutant beta-amyloid precursor protein (APP) lowered their brain levels of Abeta42. In cultured cells, the decrease in Abeta42 secretion was accompanied by an increase in the Abeta(1-38) isoform, indicating that NSAIDs subtly alter gamma-secretase activity without significantly perturbing other APP processing pathways or Notch cleavage. Our findings suggest that NSAIDs directly affect amyloid pathology in the brain by reducing Abeta42 peptide levels independently of COX activity and that this Abeta42-lowering activity could be optimized to selectively target the pathogenic Abeta42 species.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Peptide Fragments/metabolism , Sulindac/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/etiology , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspartic Acid Endopeptidases , Brain/metabolism , CHO Cells , Cricetinae , Disease Models, Animal , Endopeptidases/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Ibuprofen/pharmacology , Indomethacin/pharmacology , Mass Spectrometry , Mice , Mice, Transgenic , Prostaglandin-Endoperoxide Synthases/metabolism , Sulindac/pharmacology , Tumor Cells, Cultured
10.
Science ; 294(5549): 2179-81, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11679632

ABSTRACT

ErbB-4 is a transmembrane receptor tyrosine kinase that regulates cell proliferation and differentiation. After binding of its ligand heregulin (HRG) or activation of protein kinase C (PKC) by 12-O-tetradecanoylphorbol-13-acetate (TPA), the ErbB-4 ectodomain is cleaved by a metalloprotease. We now report a subsequent cleavage by gamma-secretase that releases the ErbB-4 intracellular domain from the membrane and facilitates its translocation to the nucleus. gamma-Secretase cleavage was prevented by chemical inhibitors or a dominant negative presenilin. Inhibition of gamma-secretase also prevented growth inhibition by HRG. gamma-Secretase cleavage of ErbB-4 may represent another mechanism for receptor tyrosine kinase-mediated signaling.


Subject(s)
Cell Nucleus/metabolism , Endopeptidases/metabolism , ErbB Receptors/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , COS Cells , Carbamates/pharmacology , Cell Division/drug effects , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Dipeptides/pharmacology , ErbB Receptors/chemistry , Fatty Acids, Unsaturated/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Mice , Molecular Sequence Data , Mutation , Neuregulin-1/pharmacology , Presenilin-1 , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Receptor, ErbB-4 , Recombinant Fusion Proteins/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation , Tumor Cells, Cultured
11.
Drug Discov Today ; 6(20): 1049-1055, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11590033

ABSTRACT

The basis for therapeutic strategies targeting the amyloid-beta protein (Abeta) has come from studies showing that accumulation and aggregation of the Abeta within the brain is likely to cause Alzheimer's disease (AD). Along with an ever-increasing understanding of Abeta metabolism, many potential therapeutic strategies aimed at altering Abeta metabolism have emerged. Among the more intriguing targets for therapy are enzymes involved in cholesterol homeostasis, because it has been found that altering cholesterol can influence Abeta metabolism in experimental model systems, and that cholesterol-lowering agents, specifically HMG-CoA reductase inhibitors, could reduce the incidence of AD. It is likely that cholesterol influences Abeta metabolism in several ways, including altering Abeta production and perhaps altering Abeta deposition and clearance. Thus, pharmacological modulation of cholesterol levels could provide a relatively safe means to reduce Abeta accumulation in the brain, and thereby prevent or slow the development of AD.

12.
J Biol Chem ; 276(42): 38563-9, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11504726

ABSTRACT

Presenilin 1 (PS1) is linked with Alzheimer's disease but exhibits functional roles regulating growth and development. For instance, PS1 binds to beta-catenin and modulates beta-catenin signaling. In the current study, we observed that knockout of PS1 inhibited beta-catenin-mediated transcription by 35%, as shown by a luciferase reporter driven by the hTcf-4 promoter. Overexpressing wild-type PS1 increased beta-catenin-mediated transcription by 37.5%, and overexpressing PS1 with mutations associated with Alzheimer's disease decreased beta-catenin-mediated transcription by 66%. To examine whether regulation of beta-catenin by PS1 requires phosphorylation by glycogen synthase kinase 3beta (GSK 3beta), we examined whether inhibiting GSK 3beta activity overcomes the inhibition of beta-catenin transcription induced by mutant PS1 constructs. Cells expressing wild-type or mutant PS1 were treated with LiCl, which inhibits GSK 3beta, or transfected with beta-catenin constructs that lack the GSK 3beta phosphorylation sites. Neither treatment overcame PS1-mediated inhibition of beta-catenin signaling, suggesting that regulation of beta-catenin by PS1 was not affected by the activity of GSK 3beta. To investigate how PS1 might regulate beta-catenin signaling, we determined whether PS1 interacts with other elements of the beta-catenin signaling cascade, such as the Tcf-4 transcription factor. Coimmunoprecipitation studies showed binding of PS1 and hTcf-4, and examining nuclear isolates indicated that nuclear hTcf-4 was decreased in cells expressing mutant PS1. These data show that PS1 interacts with multiple components of the beta-catenin signaling cascade and suggest that PS1 regulates beta-catenin in a manner independent of GSK 3beta activity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Trans-Activators , Transcription, Genetic , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Immunoblotting , Immunohistochemistry , Lithium Chloride/pharmacology , Luciferases/metabolism , Mice , Mice, Knockout , Mutation , Plasmids/metabolism , Precipitin Tests , Presenilin-1 , Promoter Regions, Genetic , Protein Binding , Signal Transduction , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription Factors/metabolism , beta Catenin
13.
J Neurosci Methods ; 108(2): 171-9, 2001 Jul 30.
Article in English | MEDLINE | ID: mdl-11478976

ABSTRACT

Evidence gathered over the last two decades suggests that beta amyloid (Abeta), the predominant proteinaceous component of senile plaques, plays an early and critical role in the etiology and pathogenesis of Alzheimer's disease (AD). Thus, it is reasonable to hypothesize that compounds capable of reducing the accumulation of Abeta may be of value therapeutically. Additionally, compounds that influence Abeta accumulation may be useful as tools to further dissect the cellular pathways that regulate Abeta production and accumulation. To screen for compounds that affect Abeta levels, we have established high throughput, cell-based assays capable of the sensitive and selective detection of Abeta40 in parallel with the more amyloidogenic form of the peptide, Abeta42. To validate the approach, we examined the effects of several compounds previously identified to influence Abeta accumulation. Analysis of peptide accumulation following treatment with these compounds showed results similar to those previously published. Currently, we are using this assay to screen drugs that have already received FDA approval for the treatment of other diseases and over-the-counter natural product extracts. If compounds such as these can be identified that lower Abeta in the brain, they may represent one of the fastest and most cost effective methods to therapy.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/drug effects , Biological Assay/methods , Cells, Cultured/drug effects , Drug Evaluation, Preclinical/methods , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Biological Assay/instrumentation , CHO Cells/drug effects , CHO Cells/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured/metabolism , Cricetinae , Culture Media, Conditioned/pharmacology , Drug Evaluation, Preclinical/instrumentation , Drug-Related Side Effects and Adverse Reactions/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , Peptide Fragments/analysis , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Reproducibility of Results , Toxicity Tests/instrumentation , Toxicity Tests/methods
14.
Biochim Biophys Acta ; 1537(1): 58-62, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11476963

ABSTRACT

In order to develop transgenic animal models that selectively overexpress various Abeta peptides, we have developed a novel expression system that selectively expresses Abeta40 or Abeta42 in the secretory pathway. This system utilizes fusion constructs in which the sequence encoding the 23-amino-acid ABri peptide at the carboxyl terminus of the 266-amino-acid type 2 transmembrane protein BRI is replaced with a sequence encoding either Abeta40 or Abeta42. Constitutive processing of the resultant BRI-Abeta fusion proteins in transfected cells results in high-level expression and secretion of the encoded Abeta peptide. Significantly, expression of Abeta42 from the BRI-Abeta42 construct resulted in no increase in secreted Abeta40, suggesting that the majority of Abeta42 is not trimmed by carboxypeptidase to Abeta40 in the secretory pathway.


Subject(s)
Amyloid beta-Peptides/genetics , Amyloid/genetics , Recombinant Fusion Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cells, Cultured , Humans , Membrane Glycoproteins , Membrane Proteins , Mice , Models, Molecular , Peptide Fragments/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Transfection
15.
J Neurosci ; 21(14): 4987-95, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438574

ABSTRACT

Alzheimer's disease (AD) is a disorder of two pathologies: amyloid plaques, the core of which is a peptide derived from the amyloid precursor protein (APP), and neurofibrillary tangles composed of highly phosphorylated tau. Protein kinase C (PKC) is known to increase non-amyloidogenic alpha-secretase cleavage of APP, producing secreted APP (sAPPalpha), and glycogen synthase kinase (GSK)-3beta is known to increase tau phosphorylation. Both PKC and GSK-3beta are components of the wnt signaling cascade. Here we demonstrate that overexpression of another member of this pathway, dishevelled (dvl-1), increases sAPPalpha production. The dishevelled action on APP is mediated via both c-jun terminal kinase (JNK) and protein kinase C (PKC)/mitogen-activated protein (MAP) kinase but not via p38 MAP kinase. These data position dvl-1 upstream of both PKC and JNK, thereby explaining the previously observed dual signaling action of dvl-1. Furthermore, we show that human dvl-1 and wnt-1 also reduce the phosphorylation of tau by GSK-3beta. Therefore, both APP metabolism and tau phosphorylation are potentially linked through wnt signaling.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Aspartic Acid Endopeptidases , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Dishevelled Proteins , Endopeptidases/metabolism , Gene Expression , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , JNK Mitogen-Activated Protein Kinases , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mutation , Phosphoproteins/genetics , Phosphoproteins/pharmacology , Phosphorylation/drug effects , Proteins/genetics , Proteins/metabolism , Proteins/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Wnt Proteins , Wnt1 Protein , tau Proteins/genetics , tau Proteins/metabolism
16.
Trends Mol Med ; 7(6): 264-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11378516

ABSTRACT

Studies demonstrating that accumulation and aggregation of the amyloid beta protein (Abeta) within the brain is likely to cause Alzheimer's disease (AD) have provided the rationale for therapeutic strategies aimed at influencing Abeta production, aggregation and clearance. gamma-secretase catalyzes the final cleavage that releases the Abeta from its precursor; therefore, it is a potential therapeutic target for the treatment of AD. Recent data show that the polytopic membrane proteins presenilin 1 and presenilin 2 are either catalytic components or essential co-factors of a membrane-bound proteolytic complex that possesses gamma-secretase activity. Although recent findings demonstrating that gamma-secretase inhibitors bind directly to presenilins (PSs) further support a catalytic role for PSs in gamma-secretase cleavage, additional studies are still needed to clarify the role of PSs in gamma-secretase cleavage and the use of targeting PSs to reduce Abeta production.


Subject(s)
Alzheimer Disease/therapy , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , Catalysis , Endopeptidases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Models, Biological , Presenilin-1 , Presenilin-2
17.
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
18.
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
19.
Ann Neurol ; 48(5): 806-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11079548

ABSTRACT

We describe 3 new families affected by Alzheimer's disease with spastic paraparesis. In affected individuals, including the earliest known patient with this clinical syndrome, neuropathological examination revealed large "cotton wool" plaques similar to those we have previously described in a Finnish family. In the families in which DNA was available, presenilin-1 mutations were observed. Transfection of cells with these mutant genes caused exceptionally large increases in secreted Abeta42 levels. Furthermore, brain tissue from individuals with this syndrome had very high amyloid-beta concentrations. These findings define the molecular pathogenesis of an important subgroup of Alzheimer's disease and have implications for the pathogenesis of the disease in general.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/analysis , Paraparesis, Spastic/genetics , Paraparesis, Spastic/pathology , Alzheimer Disease/complications , Finland , Humans , Mutation/genetics , Paraparesis, Spastic/complications
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...