Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 134: 104636, 2020 02.
Article in English | MEDLINE | ID: mdl-31629891

ABSTRACT

Abnormal tau hyperphosphorylation and its aggregation into neurofibrillary tangles are a hallmark of tauopathies, neurodegenerative disorders that include Alzheimer's disease (AD). Active and passive Tau-immunotherapy has been proposed as a therapeutic approach to AD with mixed results. One of the limitations of active immunotherapy may be associated with the mediocre immunogenicity of vaccines that are not inducing therapeutically potent titers of antibodies. The aim of this study was to test the efficacy of an anti-tau vaccine, AV-1980R/A composed of N terminal peptide of this molecule fused with an immunogenic MultiTEP platform and formulated in a strong adjuvant, AdvaxCpG in a Tg4510 mouse model of tauopathy. Experimental mice were immunized with AV-1980R/A and a control group of mice were injected with adjuvant only. Nontransgenic and tetracycline transactivator (tTA) transgenic littermates were included as baseline controls to contrast with the tau phenotype. Active immunization with AV-1980R/A induced very strong anti-tau humoral immune responses in both nontransgenic and transgenic mice with evidence of IgG in brains of AV-1980R/A vaccinated mice. These experimental animals displayed an improvement in short-term memory during a novel object recognition test. However, impairments in other behavioral tasks were not prevented by AV-1980R/A vaccinations. At the same time, high titers of anti-tau antibodies reduced hyperphosphorylated pSer396 tau but did not lower the level of other phosphorylated tau species in the brains of AV-1980R/A vaccinated mice. These data indicate that active immunotherapy with an N-terminal Tau epitope was only partially effective in improving cognition and reducing pathology in the stringent Tg4510 mouse model of tauopathy.


Subject(s)
Alzheimer Vaccines , Immunogenicity, Vaccine/immunology , Tauopathies , Vaccination , tau Proteins/immunology , Animals , Antibody Formation , Disease Models, Animal , Epitopes/immunology , Memory , Mice , Mice, Transgenic
2.
J Neuroimmunol ; 223(1-2): 104-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20451261

ABSTRACT

Anti-amyloid immunotherapy has been proposed as an appropriate therapeutic approach for Alzheimer's disease (AD). Significant efforts have been made towards the generation and assessment of antibody-based reagents capable of preventing and clearing amyloid aggregates as well as preventing their synaptotoxic effects. In this study, we selected a novel set of human anti-amyloid-beta peptide 1-42 (Abeta1-42) recombinant monoclonal antibodies in a single chain fragment variable (scFv) and a single-domain (VH) format. We demonstrated that these antibody fragments recognize in a specific manner amyloid-beta deposits in APP/Tg mouse brains, inhibit toxicity of oligomeric Abeta1-42 in neuroblastoma cell cultures in a concentration-dependent manner and reduced amyloid deposits in APP/Tg2576 mice after intracranial administration. These antibody fragments recognize epitopes in the middle/C-terminus region of Abeta, which makes them strong therapeutic candidates due to the fact that most of the Abeta species found in the brains of AD patients display extensive N-terminus truncations/modifications.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/chemistry , Bacteriophage M13/immunology , Epitopes/immunology , Immunoglobulin Heavy Chains/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Library , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Bacteriophage M13/chemistry , Bacteriophage M13/genetics , Binding Sites, Antibody/genetics , Cell Line, Tumor , Epitopes/genetics , Epitopes/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics
3.
Gene Ther ; 17(2): 261-71, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19865176

ABSTRACT

Recently, we reported that a DNA vaccine, composed of three copies of a self B cell epitope of amyloid-beta (Abeta(42)) and the foreign T-cell epitope, Pan DR epitope (PADRE), generated strong anti-Abeta immune responses in wild-type and amyloid precursor protein transgenic animals. Although DNA vaccines have several advantages over peptide-protein vaccines, they induce lower immune responses in large animals and humans compared with those in mice. The focus of this study was to further enhance anti-Abeta(11) immune responses by developing an improved DNA vaccination protocol of the prime-boost regimen, in which the priming step would use DNA and the boosting step would use recombinant protein. Accordingly, we generated DNA and recombinant protein-based epitope vaccines and showed that priming with DNA followed by boosting with a homologous recombinant protein vaccine significantly increases the anti-Abeta antibody responses and do not change the immunoglobulin G1 (IgG1) profile of humoral immune responses. Furthermore, the antibodies generated by this prime-boost regimen were long-lasting and possessed a higher avidity for binding with an Abeta(42) peptide. Thus, we showed that a heterologous prime-boost regimen could be an effective protocol for developing a potent Alzheimer's disease (AD) vaccine.


Subject(s)
Amyloid beta-Peptides/immunology , Immunization, Secondary , Vaccines, DNA/immunology , Vaccines, Subunit/immunology , Animals , Antibody Affinity , Chemokine CCL22/immunology , Epitopes, T-Lymphocyte/immunology , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/metabolism , Malaria Vaccines/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/immunology , Vaccines, Synthetic/immunology
4.
J Neuroimmunol ; 213(1-2): 39-46, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19545911

ABSTRACT

N-truncated and N-modified forms of amyloid beta (Abeta) peptide are found in diffused and dense core plaques in Alzheimer's disease (AD) and Down's syndrome patients as well as transgenic mouse models of AD. Although the pathological significance of these shortened forms Abeta is not completely understood, previous studies have demonstrated that these peptides are significantly more resistant to degradation, aggregate more rapidly in vitro and exhibit similar or, in some cases, increased toxicity in hippocampal neuronal cultures compared to the full length peptides. In the present study we further investigated the mechanisms of toxicity of one of the most abundant N-truncated/modified Abeta peptide bearing amino-terminal pyroglutamate at position 3 (AbetaN3(pE)). We demonstrated that AbetaN3(pE) oligomers induce phosphatidyl serine externalization and membrane damage in SH-SY5Y cells. Also, we produced AbetaN3(pE)-specific polyclonal antibodies in rabbit and identified an immunodominant epitope recognized by anti-AbetaN3(pE) antibodies. Our results are important for developing new immunotherapeutic compounds specifically targeting AbetaN3(pE) aggregates since the most commonly used immunogens in the majority of vaccines for AD have been shown to induce antibodies that recognize the N-terminal immunodominant epitope (EFRH) of the full length Abeta, which is absent in N-amino truncated peptides.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Antibodies/immunology , Immunodominant Epitopes/immunology , Pyrrolidonecarboxylic Acid/immunology , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Alzheimer Vaccines/chemistry , Alzheimer Vaccines/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Animals , Antibodies/pharmacology , Antibody Specificity/immunology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Humans , Molecular Structure , Molecular Weight , Peptides/chemistry , Peptides/immunology , Peptides/toxicity , Phosphatidylserines/metabolism , Protein Structure, Tertiary/physiology , Pyrrolidonecarboxylic Acid/chemistry , Rabbits
5.
Gene Ther ; 15(1): 61-4, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17972923

ABSTRACT

The ideal immunological target for cancer vaccine development would meet the criteria of tumor specificity, immunogenicity and vital dependency of the tumor on the functional activities of the antigenic target so as to avoid antigenic loss by mutation. Given that at face value the brother of regulator of imprinted sites (BORIS) transcription factor meets these criteria, we have developed a mutant variant of this molecule (mBORIS) that lacks tumorigenic ability, while retaining immunogenic epitopes that elicits responses against histologically irrelevant tumor cells. Here we compared vaccine strategies employing as an immunogen either mBORIS recombinant protein formulated in a strong Th1-type adjuvant, QuilA or DNA encoding this immunogen along with plasmids expressing interleukin (IL)12/IL18 molecular adjuvants. In both groups of vaccinated mice induction of tumor-specific immunity (antibody response, T-cell proliferation, cytokine production, T-cell cytotoxicity) as well as ability to inhibit growth of the aggressive breast cancer cell line and to prolong survival of vaccinated animals have been tested. We determined that DNA, but not recombinant protein vaccine, induced potent Th1-like T-cell recall responses that significantly inhibited tumor growth and prolongs the survival of vaccinated mice. These studies demonstrate that DNA immunization is superior to recombinant protein strategy and provide a clear guidance for clinical development of a cancer vaccine targeting what appears to be a universal tumor antigen.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Immunotherapy/methods , Vaccines, DNA/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Breast Neoplasms/immunology , Cancer Vaccines/genetics , Cell Line, Tumor , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-18/genetics , Interleukin-4/immunology , Mice , Mutation , Neoplasm Transplantation , Quillaja Saponins , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Saponins/administration & dosage , Th1 Cells/immunology , Treatment Outcome , Vaccines, DNA/genetics
6.
Mol Immunol ; 45(4): 881-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17889938

ABSTRACT

Active and passive immunotherapy targeted at the amyloid-beta (Abeta) peptide has been proposed as therapeutic approach against Alzheimer's disease (AD), and efforts towards the generation and application of antibody-based reagents that are capable of preventing and clearing amyloid aggregates are currently under active investigation. Previously, we selected and characterized a new anti-Abeta1-42 phage-displayed scFv antibody, designated clone b4.4, using a non-immune human scFv antibody library and demonstrated that a peptide based on the sequence of the Ig heavy chain (VH) complementarity-determining region (HCDR3) of this antibody fragment bound to Abeta1-42)and had neuroprotective potential against Abeta1-42 mediated neurotoxicity in rat hippocampal cultured neurons. In the present study, using novel computational methods and in vitro experiments we demonstrated that b4.4 binds to the central region of Abeta1-42. We also demonstrated that this scFv antibody binds to Abeta-derived diffusible ligands (ADDLs) and neutralizes the toxicity of both fibrillar and oligomeric forms of Abeta1-42 tested in vitro in SH-SY5Y cell cultures.


Subject(s)
Amyloid beta-Peptides/pharmacology , Complementarity Determining Regions/pharmacology , Immunoglobulin Fragments/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Cell Line, Tumor , Cell Survival/drug effects , Complementarity Determining Regions/chemistry , Epitope Mapping , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Heavy Chains/chemistry , Models, Molecular , Neuroprotective Agents/chemistry , Oligopeptides/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Binding
7.
Neurobiol Aging ; 28(12): 1821-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17052803

ABSTRACT

Alzheimer disease (AD) is the most prominent cause of dementia in the elderly. To determine changes in the AD brain that may mediate the transition into dementia, the gene expression of approximately 10,000 full-length genes was compared in mild/moderate dementia cases to non-demented controls that exhibited high AD pathology. Including this latter group distinguishes this work from previous studies in that it allows analysis of early cognitive loss. Compared to non-demented high-pathology controls, the hippocampus of AD cases with mild/moderate dementia had increased gene expression of the inflammatory molecule major histocompatibility complex (MHC) II, as assessed with microarray analysis. MHC II protein levels were also increased and inversely correlated with cognitive ability. Interestingly, the mild/moderate AD dementia cases also exhibited decreased number of T cells in the hippocampus and the cortex compared to controls. In conclusion, transition into AD dementia correlates with increased MHC II(+) microglia-mediated immunity and is paradoxically paralleled by a decrease in T cell number, suggesting immune dysfunction.


Subject(s)
Alzheimer Disease/immunology , Brain/immunology , Cytokines/immunology , Dementia/immunology , Encephalitis/immunology , Aged , Aged, 80 and over , Female , Gene Expression Regulation/immunology , Humans
8.
Mol Cell Biol ; 25(22): 9897-909, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16260605

ABSTRACT

The human c-myc proto-oncogene, implicated in the control of many cellular processes including cell growth and apoptosis, encodes three isoforms which differ in their N-terminal region. The functions of these isoforms have never been addressed in vivo. Here, we used Drosophila melanogaster to examine their functions in a fully integrated system. First, we established that the human c-Myc protein can rescue lethal mutations of the Drosophila myc ortholog, dmyc, demonstrating the biological relevance of this model. Then, we characterized a new lethal dmyc insertion allele, which permits expression of human c-Myc in place of dMyc and used it to compare physiological activities of these isoforms in whole-organism rescue, transcription, cell growth, and apoptosis. These isoforms differ both quantitatively and qualitatively. Most remarkably, while the small c-MycS form truncated for much of its N-terminal trans-activation domain efficiently rescued viability and cell growth, it did not induce detectable programmed cell death. Our data indicate that the main functional difference between c-Myc isoforms resides in their apoptotic properties and that the N-terminal region, containing the conserved MbI motif, is decisive in governing the choice between growth and death.


Subject(s)
Apoptosis , Gene Expression Regulation , Proto-Oncogene Proteins c-myc/chemistry , Alleles , Amino Acid Motifs , Animals , Cell Cycle , Cell Proliferation , Cloning, Molecular , Drosophila melanogaster , Exons , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mitosis , Models, Genetic , Mutation , Plasmids/metabolism , Protein Isoforms , Protein Structure, Tertiary , Proto-Oncogene Mas , RNA/chemistry , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Temperature , Transcription, Genetic , Transgenes
9.
Neurobiol Dis ; 8(6): 1006-16, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11741396

ABSTRACT

Recent studies support the activation of apoptotic pathways in the Alzheimer's disease (AD) brain. Neurons committed to apoptosis may do so by either activation of a receptor-mediated pathway employing caspase-8 or through an alternative mitochondrial pathway involving oxidative stress. In the present study, the role of caspase-8 in the AD brain was examined by designing a caspase-cleavage site-directed antibody to one of the active fragments of caspase-8. In vitro analysis with this antibody, termed CASP-8p18, demonstrated that it recognized the active 18-kDa fragment of caspase-8 but not the precursor protein. In vivo immunohistochemical analysis using hippocampal tissue sections from AD or aged-matched control brains demonstrated CASP-8p18 immunolabeling of neurons in all AD cases, whereas little staining was observed in controls. These results were confirmed using a commercially available antibody that, like the CASP-8p18 antibody reacts only with the 18-kDa fragment of caspase-8 and not full-length caspase-8. As with CASP-8p18 antibody, the commercial antibody-labeled neurons in all AD cases, while showing a relative paucity of staining in representative control cases. Labeling of CASP-8p18 within tangle-bearing neurons was observed in double-labeling studies with AT8 or PHF-1, both markers for neurofibrillary tangles (NFTs). In addition, using a caspase-cleavage site-directed antibody that recognizes cleavage products of caspase-3 showed colocalization of this antibody with the CASP-8p18 antibody within NFTs. These results suggest a role for caspase-8 and the receptor-mediated apoptotic pathway as a mechanism leading to the activation of caspase-3 within neurons of the AD brain.


Subject(s)
Alzheimer Disease/enzymology , Apoptosis/physiology , Caspases/metabolism , Entorhinal Cortex/enzymology , Hippocampus/enzymology , Neurons/enzymology , Aged , Aging/metabolism , Aging/pathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity , Blotting, Western , Carrier Proteins/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/immunology , Cell Compartmentation/immunology , Dogs , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Female , HeLa Cells , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Immunohistochemistry , Male , Microfilament Proteins/metabolism , Middle Aged , Neurofibrillary Tangles/enzymology , Neurofibrillary Tangles/pathology , Neurons/pathology
10.
Neurobiol Dis ; 8(2): 252-65, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11300721

ABSTRACT

To study the link between beta-amyloid (Abeta) and neuroinflammation, we examined the levels of complement as a function of age and extent of Abeta deposition in Down Syndrome (DS) brain. C1q, the first component of the complement cascade, was visualized using immunohistochemistry in the frontal, entorhinal cortex, and hippocampus of 12 DS ranging from 31 to 69 years of age. C1q was consistently associated with thioflavine-S positive Abeta plaques in DS brain and increased with more extensive age-dependent Abeta deposition. In contrast, little or no C1q labeling was associated with diffuse or thioflavine-S negative Abeta deposits. Neurons in the hippocampus and entorhinal cortex, but less frequently in frontal cortex, were C1q positive in DS cases with sufficient neuropathology to have a diagnosis of Alzheimer's disease. C1q-positive neurons were associated with activated microglia. These results provide evidence for Abeta-mediated inflammatory factors contributing to the rapid accumulation of neuropathology in DS brain.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/metabolism , Brain/immunology , Complement C1q/metabolism , Down Syndrome/immunology , Neurons/immunology , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Benzothiazoles , Brain/metabolism , Brain/pathology , Complement C1q/immunology , Down Syndrome/metabolism , Down Syndrome/pathology , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/pathology , Female , Humans , Immunohistochemistry , Male , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Middle Aged , Neurofibrillary Tangles/immunology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Thiazoles/metabolism
11.
Neuroreport ; 12(4): 839-43, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11277593

ABSTRACT

Although considerable research has shown a role for peroxisome proliferator-activated receptors (PPAR) in adipose differentiation and in the regulation of inflammation, little is known about its possible functions in neurons. We investigated the role of PPARgamma in primary cultures of cortical neurons and human neuroblastoma SH-SYSY cells. Incubation of cortical neurons with the specific PPARgamma ligand 15-Deoxy-delta12,14-prostaglandin J2 (15d-PGJ2) induced morphological changes including neurite degeneration and nuclear condensation that were consistent with neurons dying by apoptosis. The morphological changes associated with incubation of cortical neurons with 15d-PGJ2 were prevented following pretreatment of neurons with the general caspase inhibitor, Z-VAD. These results highlight a novel role for PPARgamma in neurons and suggest that unwarranted activation of PPARgamma may contribute to the neuronal apoptosis associated with certain neurodegenerative disorders including Alzheimer's disease (AD).


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Immunologic Factors/pharmacology , Neurons/cytology , Prostaglandin D2/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Cerebral Cortex/cytology , Humans , Neuroblastoma , Neurons/physiology , Poly(ADP-ribose) Polymerases/metabolism , Prostaglandin D2/analogs & derivatives , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
12.
EMBO J ; 20(4): 802-11, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11179224

ABSTRACT

Hox genes encoding homeodomain transcriptional regulators are known to specify the body plan of multicellular organisms and are able to induce body plan transformations when misexpressed. These findings led to the hypothesis that duplication events and misexpression of Hox genes during evolution have been necessary for generating the observed morphological diversity found in metazoans. It is known that overexpressing Antennapedia (Antp) in the head induces antenna-to-leg as well as head-to-thorax transformation and eye reduction. At present, little is known about the exact molecular mechanism causing these phenotypes. The aim of this study is to understand the basis of inhibition of eye development. We demonstrate that Antp represses the activity of the eye regulatory cascade. By ectopic expression, we show that Antp antagonizes the activity of the eye selector gene eyeless. Using both in vitro and in vivo experiments, we demonstrate that this inhibitory mechanism involves direct protein-protein interactions between the DNA-binding domains of EY and ANTP, resulting in mutual inhibition.


Subject(s)
Drosophila/embryology , Eye/embryology , Homeodomain Proteins/physiology , Nuclear Proteins , Transcription Factors , Animals , Antennapedia Homeodomain Protein , Apoptosis , Drosophila/genetics , Drosophila Proteins , Embryonic Development , Eye/cytology , Homeodomain Proteins/metabolism , Protein Binding
13.
Am J Pathol ; 158(1): 189-98, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11141492

ABSTRACT

Although evidence suggests that neurofibrillary tangles (NFTs) and neuronal cell loss are prominent features of Alzheimer's disease (AD), the relationship between the two remains unknown. In the present study, the relationship between the activation of apoptotic mechanisms and NFT formation in AD was investigated using a caspase-cleavage site-directed antibody to fodrin, an abundant neuronal cytoskeleton protein. This antibody recognized cleavage products of fodrin after digestion by caspase-3, but did not recognize full-length fodrin. In vitro analysis of this fodrin caspase-cleavage product (CCP) antibody demonstrates that it is a specific probe for the detection of apoptotic but not necrotic pathways in cultured neurons. To determine whether caspases cleave fodrin in vivo, tissue sections from controls and AD were immunostained for fodrin (CCPs). Although no staining was observed in control cases, labeling of neurons was observed in the hippocampus of all AD cases, which increased as a function of disease progression. To determine a possible relationship between caspase activation and NFT formation, double-labeling experiments with fodrin CCP and PHF-1 were performed. Co-localization of these markers was observed in many neurons, and quantitative analysis showed that as the extent of NFT formation increased, there was a significant corresponding increase in fodrin CCP immunolabeling (r = 0.84). Taken together, these results provide evidence for the activation of apoptotic mechanisms in neurons in the AD brain and suggest that there is an association between NFT formation and the activation of apoptotic pathways in AD.


Subject(s)
Alzheimer Disease/metabolism , Caspases/metabolism , Neurofibrillary Tangles/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Apoptosis , Blotting, Western , Brain/metabolism , Brain/pathology , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell-Free System , Cells, Cultured , Dogs , Enzyme Activation , Female , Hippocampus/chemistry , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Microscopy, Confocal , Middle Aged , Neurons/chemistry , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
14.
J Neurobiol ; 46(2): 73-88, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11153010

ABSTRACT

A role for the Pax-6 homologue eyeless in adult Drosophila brain development and function is described. eyeless expression is detected in neurons, but not glial cells, of the mushroom bodies, the medullar cortex, the lateral horn, and the pars intercerebralis. Furthermore, severe defects in adult brain structures essential for vision, olfaction, and for the coordination of locomotion are provoked by two newly isolated mutations of Pax-6/eyeless that result in truncated proteins. Consistent with the morphological lesions, we observe defective walking behavior for these eyeless mutants. The implications of these data for understanding postembryonic brain development and function in Drosophila are discussed.


Subject(s)
Brain/growth & development , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila/growth & development , Homeodomain Proteins/metabolism , Age Factors , Alleles , Animals , Brain/cytology , Brain/metabolism , DNA Mutational Analysis/statistics & numerical data , Drosophila/cytology , Drosophila/metabolism , Eye Proteins , Gait Disorders, Neurologic/genetics , Gait Disorders, Neurologic/pathology , Gait Disorders, Neurologic/physiopathology , Genetic Complementation Test , Models, Animal , Mutation/physiology , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Olfaction Disorders/genetics , Olfaction Disorders/pathology , Olfaction Disorders/physiopathology , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/growth & development , Optic Lobe, Nonmammalian/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , Repressor Proteins , Vision Disorders/genetics , Vision Disorders/pathology , Vision Disorders/physiopathology
15.
Genes Dev ; 14(22): 2906-17, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11090137

ABSTRACT

The origins of specificity in gene expression are a central concern in understanding developmental control. Mediator protein complexes regulate transcriptional initiation, acting as modular adaptors linking specific transcription factors to core RNA polymerase II. Here, we identified the Drosophila homologs of 23 human mediator genes and mutations of two, dTRAP240 and of dTRAP80 (the putative fly homolog of yeast SRB4). Clonal analysis indicates a general role for dTRAP80 necessary for cell viability. The dTRAP240 gene is also essential, but cells lacking its function are viable and proliferate normally. Clones reveal localized developmental activities including a sex comb cell identity function. This contrasts with the ubiquitous nuclear accumulation of dTRAP240 protein in imaginal discs. Synergistic genetic interactions support shared developmental cell and segment identity functions of dTRAP240 and dTRAP80, potentially within a common complex. Further, they identify the homeotic Sex combs reduced product, required for the same cell/tissue identities, as a functional partner of these mediator proteins.


Subject(s)
Body Patterning , Drosophila Proteins , Drosophila/embryology , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Cloning, Molecular , Genes, Homeobox , Genes, Insect , Humans , Insect Proteins , Molecular Sequence Data , Sequence Homology, Amino Acid , Species Specificity
16.
Amyloid ; 7(3): 153-65, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11019856

ABSTRACT

Amyloid precursor protein (A betaPP) processing results in generation of amyloid beta peptide (A beta) which deposits in the brain parenchyma and cerebrovasculature of patients with Alzheimer's disease (AD). Evidence that the vascular deposits derive in part from A betaPP fragments originating from activated platelets includes findings that individuals who have had multiple small strokes have a higher prevalence of AD compared to individuals who have taken anti-platelet drugs. Thus, determination of whether platelet A betaPP fragments are capable of traversing the blood-brain barrier (BBB) is critical. We have established that activated platelets from patients with AD retain more surface transmembrane-bound A betaPP (mA betaPP) than control platelets. We report here that this mA betaPP can be cleaved to A beta-containing fragments which pass through a novel BBB model system. This model utilizes human BBB endothelial cells (BEC) isolated from brains of patients with AD. These BEC, after exposure to activated platelets which have been surface-labeled with fluorescein and express surface-retained mA betaPP, cleave fluorescein-tagged surface proteins, including mA betaPP, resulting in passage to the BEC layer The data confirm that BEC contribute to processing of platelet-derived mA betaPP and show that the processing yields A beta containing fragments which could potentially contribute to cerebrovascular A beta deposition.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Blood-Brain Barrier/physiology , Endothelium, Vascular/metabolism , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Brain/blood supply , Brain/pathology , Cells, Cultured , Endothelium, Vascular/pathology , Female , Fluorescein-5-isothiocyanate/analysis , Fluorescent Dyes/analysis , Humans , Male , Microscopy, Confocal , Middle Aged , Platelet Activation
17.
Biochemistry ; 39(20): 5988-94, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10821670

ABSTRACT

Despite significant progress in the elucidation of the genetic basis of early-onset familial Alzheimer's disease (AD), the etiology of sporadic cases remains elusive. Although certain genetic loci play a role in conferring susceptibility in some sporadic AD cases, it is likely that the etiology is multifactorial; hence, the majority of cases cannot be attributed to genetic factors alone, indicating that environmental factors may modulate the onset and/or progression of the disease. Head injury and infectious agents are environmental factors that have been periodically implicated, but no plausible mechanisms have been clearly identified. With regard to infectious agents, speculation has often centered on the neurotropic herpes viruses, with herpes simplex virus 1 (HSV1) considered a likely candidate. We report that an internal sequence of HSV1 glycoprotein B (gB) is homologous to the carboxyl-terminal region of the A beta peptide that accumulates in diffuse and neuritic plaques in AD. Synthetic peptides were generated and the biophysical and biological properties of the viral peptide compared to those of A beta. Here we show that this gB fragment forms beta-pleated sheets, self-assembles into fibrils that are thioflavin-positive and ultrastructurally indistinguishable from A beta, accelerates the formation of A beta fibrils in vitro, and is toxic to primary cortical neurons at doses comparable to those of A beta. These findings suggest a possible role for this infectious agent in the pathophysiology of sporadic cases of AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Microfibrils/metabolism , Neurotoxins/metabolism , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Sequence Homology, Amino Acid , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/toxicity , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/ultrastructure , Animals , Cell Death , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Herpesvirus 1, Human/pathogenicity , Humans , Microfibrils/chemistry , Microfibrils/ultrastructure , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotoxins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Rats , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/ultrastructure
18.
J Neurochem ; 74(6): 2331-42, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820193

ABSTRACT

Although there is considerable evidence suggesting that altered metabolism of beta-amyloid precursor protein (APP) and accumulation of its beta-amyloid fragment are key features of Alzheimer's disease (AD), the normal physiological function of APP remains elusive. We investigated the potential role of APP in neurons using the monoclonal antibody 22C11, which binds to the extracellular domain of the human, rat, or mouse APP. Exposure of cortical neurons to 22C11 induced morphological changes including neurite degeneration, nuclear condensation, and internucleosomal DNA cleavage that were consistent with neurons dying by apoptosis. Supporting a role for 22C11-mediated apoptosis occurring by binding to APP were data demonstrating that preincubation of 22C11 with either purified APP or a synthetic peptide (APP(66-81)) that contains the epitope for 22C11 significantly attenuated neuronal damage induced by 22C11. The specificity of 22C11 was further supported by data showing no apparent effects of either mouse IgG or the monoclonal antibody P2-1, which is specific for the aminoterminal end of human but not rat APP. In addition, biochemical features indicative of apoptosis were the formation of 120- and 150-kDa breakdown products of fodrin following treatment of cortical neurons with 22C11. Both the morphological and the biochemical changes induced by 22C11 were prevented following pretreatment of neurons with the general caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp(O-methyl)-fluoromethyl ketone. Prior incubation of cortical neurons with GSH ethyl ester (GEE), a cell-permeable form of GSH, resulted in complete protection from the 22C11 insult, thus implicating an oxidative pathway in 22C11-mediated neuronal degeneration. This was further supported by the observation that prior treatment of neurons with buthionine sulfoximine, an inhibitor of gamma-glutamylcysteinyl synthetase, potentiated the toxic effects of 22C11. Finally, with use of compartmented cultures of hippocampal neurons, it was also demonstrated that selective application of 22C11 caused local neuritic degeneration that was prevented by the addition of GEE to the neuritic compartment. Thus, the binding of a monoclonal antibody to APP initially triggers neurite degeneration that is followed by caspase-dependent apoptosis in neuronal cultures and illustrates a novel property of this protein in neurons that may contribute to the profound neuronal cell death associated with AD.


Subject(s)
Amyloid beta-Protein Precursor/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/immunology , Neurons/ultrastructure , Alzheimer Disease/pathology , Animals , Carrier Proteins/metabolism , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Hippocampus/cytology , Microfilament Proteins/metabolism , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neurites/immunology , Neurites/pathology , Neurons/enzymology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
19.
Neurobiol Aging ; 20(3): 297-304, 1999.
Article in English | MEDLINE | ID: mdl-10588577

ABSTRACT

In transgenic models of Alzheimer's disease (AD) neuronal loss has not been widely observed. The loss of neurons in AD may be due to chronic activation of complement (C') by beta-amyloid (A beta). A beta has been shown to activate C' by binding to a site on the C1q A-chain. The mouse A-chain sequence differs significantly from human, and a peptide based on the mouse A-chain sequence was ineffective at blocking activation of C' by A beta in contrast to the inhibition seen with the human peptide. Comparison of mouse and human serum showed that human C' was activated more effectively by A beta than was mouse C'. Therefore, additional genetic manipulations may be necessary to replicate in the murine model the inflammation and neurodegeneration that occur in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Complement Activation/physiology , Complement C1q/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/pharmacology , Animals , Binding Sites/physiology , Complement Activation/drug effects , Complement C1q/chemistry , Complement C1q/pharmacology , Dose-Response Relationship, Drug , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Peptide Fragments/pharmacology , Protein Structure, Quaternary , Species Specificity
20.
Amyloid ; 6(3): 157-64, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10524279

ABSTRACT

The amyloid beta-protein (A beta) pathologically accumulates in cerebral vascular and senile plaque deposits in the brains of patients with Alzheimer's disease (AD) and related disorders including hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). The cerebrovascular deposits are accompanied by degeneration and eventual loss of smooth muscle cells in cerebral vessel wall. Similarly, we have shown that pathogenic forms of A beta cause cell death in cultured human cerebrovascular smooth muscle (HCSM) cells in vitro. Here we show that pathogenic A beta induces a number of structural changes in HCSM cells including shrinkage of cell bodies, retraction of processes, disruption of the intracellular actin network, and nuclear condensation and fragmentation. These changes were accompanied by a number of biochemical alterations in the cells shown by in situ end labeling of nuclear DNA, proteolytic breakdown of smooth muscle cell a actin, and proteolytic activation of the proteinase caspase 3. Together, these characteristics are consistent with an apoptotic mechanism of cell death in HCSM cells in response to pathogenic A beta.


Subject(s)
Amyloid beta-Peptides/physiology , Apoptosis/physiology , Brain/blood supply , Muscle, Smooth, Vascular/pathology , Peptide Fragments/physiology , Actins/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Enzyme Activation , Humans , Hydrolysis , In Situ Nick-End Labeling , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...