Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters










Publication year range
1.
Neuropathol Appl Neurobiol ; 46(7): 673-685, 2020 12.
Article in English | MEDLINE | ID: mdl-32497293

ABSTRACT

AIMS: The deposition of amyloid-ß (Aß) peptides in the form of extracellular plaques in the brain represents one of the classical hallmarks of Alzheimer's disease (AD). In addition to 'full-length' Aß starting with aspartic acid (Asp-1), considerable amounts of various shorter, N-terminally truncated Aß peptides have been identified by mass spectrometry in autopsy samples from individuals with AD. METHODS: Selectivity of several antibodies detecting full-length, total or N-terminally truncated Aß species has been characterized with capillary isoelectric focusing assays using a set of synthetic Aß peptides comprising different N-termini. We further assessed the N-terminal heterogeneity of extracellular and vascular Aß peptide deposits in the human brain by performing immunohistochemical analyses using sporadic AD cases with antibodies targeting different N-terminal residues, including the biosimilar antibodies Bapineuzumab and Crenezumab. RESULTS: While antibodies selectively recognizing Aß1-x showed a much weaker staining of extracellular plaques and tended to accentuate cerebrovascular amyloid deposits, antibodies detecting Aß starting with phenylalanine at position 4 of the Aß sequence showed abundant amyloid plaque immunoreactivity in the brain parenchyma. The biosimilar antibody Bapineuzumab recognized Aß starting at Asp-1 and demonstrated abundant immunoreactivity in AD brains. DISCUSSION: In contrast to other studied Aß1-x -specific antibodies, Bapineuzumab displayed stronger immunoreactivity on fixed tissue samples than with sodium dodecyl sulfate-denatured samples on Western blots. This suggests conformational preferences of this antibody. The diverse composition of plaques and vascular deposits stresses the importance of understanding the roles of various Aß variants during disease development and progression in order to generate appropriate target-developed therapies.


Subject(s)
Alzheimer Disease/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Brain/metabolism , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Humans , Peptide Fragments/metabolism
2.
Adv Cancer Res ; 132: 45-71, 2016.
Article in English | MEDLINE | ID: mdl-27613129

ABSTRACT

The majority of cancer deaths are due to metastases that can occur years or decades after primary tumor diagnosis and treatment. Disseminated tumor cells (DTCs) surviving in a dormant state in target organs appear to explain the timing of this phenomenon. Knowledge on this process is important as it might provide a window of opportunity to prevent recurrences by eradicating dormant DTCs and/or by maintaining DTCs in a dormant state. Importantly, this research might offer markers of dormancy for early monitoring of metastatic relapse. However, our understanding of the mechanisms underlying the regulation of entry into and exit from dormancy is still limited and crippling any therapeutic opportunity. While cancer cell-intrinsic signaling pathways have been linked to dormancy regulation, it is likely that these pathways and the switch controlling reactivation from dormancy are regulated by microenvironmental cues. Here we review and discuss recent findings on how the microenvironment regulates cancer dormancy and raise new questions that may help advance the field.


Subject(s)
Neoplasm, Residual/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Humans
3.
Leukemia ; 29(12): 2307-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26104660

ABSTRACT

P38α/ß has been described as a tumor-suppressor controlling cell cycle checkpoints and senescence in epithelial malignancies. However, p38α/ß also regulates other cellular processes. Here, we describe a role of p38α/ß as a regulator of acute lymphoblastic leukemia (ALL) proliferation and survival in experimental ALL models. We also report first evidence that p38α/ß phosphorylation is associated with the occurrence of relapses in TEL-AML1-positive leukemia. First, in vitro experiments show that p38α/ß signaling is induced in a cyclical manner upon initiation of proliferation and remains activated during log-phase of cell growth. Next, we provide evidence that growth-permissive signals in the bone marrow activate p38α/ß in a novel avian ALL model, in which therapeutic targeting can be tested. We further demonstrate that p38α/ß inhibition by small molecules can suppress leukemic expansion and prolong survival of mice bearing ALL cell lines and primary cells. Knockdown of p38α strongly delays leukemogenesis in mice xenografted with cell lines. Finally, we show that in xenografted TEL-AML1 patients, ex vivo p38α/ß phosphorylation is associated with an inferior long-term relapse-free survival. We propose p38α/ß as a mediator of proliferation and survival in ALL and show first preclinical evidence for p38α/ß inhibition as an adjunct approach to conventional therapies.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , p38 Mitogen-Activated Protein Kinases/physiology , Adolescent , Animals , Cell Proliferation , Child , Child, Preschool , Female , Humans , Male , Mice , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
4.
Oncogene ; 33(48): 5483-90, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-24413078

ABSTRACT

The postnatal mammary gland develops extensively through cycles of proliferation, branching, involution and remodeling. We review recent advances made in the field of stress signaling pathways and its roles in mammary gland organogenesis, how they contribute to normal organ specification and homeostasis and how its subversion by oncogenes leads to cancer. We analyze stress signaling in mammary gland biology taking into account the interrelationship with the extracellular matrix and adhesion signaling during morphogenesis. By integrating the information gathered from in vivo and three dimensional in vitro organogenesis studies, we review the novel contribution of p38(SAPK), c-Jun NH2-terminal kinase and PKR-like endoplasmic reticulum kinase (PERK) signaling pathways to the timely activation of cell death, correct establishment of polarity and growth arrest and autophagy, respectively. We also review the evidence supporting that the activation of the aforementioned stress kinases maintain breast acinar structures as part of a tumor suppressive program and that its deregulation is commonplace during breast cancer initiation.


Subject(s)
Breast Neoplasms/pathology , Mammary Glands, Human , Signal Transduction/physiology , Animals , Female , Humans , Morphogenesis
5.
Oncogene ; 32(41): 4932-40, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23160380

ABSTRACT

Adhesion to the extracellular matrix (ECM) is critical for epithelial tissue homeostasis and function. ECM detachment induces metabolic stress and programmed cell death via anoikis. ECM-detached mammary epithelial cells are able to rapidly activate autophagy allowing for survival and an opportunity for re-attachment. However, the mechanisms controlling detachment-induced autophagy remain unclear. Here we uncover that the kinase PERK rapidly promotes autophagy in ECM-detached cells by activating AMP-activated protein kinase (AMPK), resulting in downstream inhibition of mTORC1-p70(S6K) signaling. LKB1 and TSC2, but not TSC1, are required for PERK-mediated inhibition of mammalian target of rapamycinin MCF10A cells and mouse embryo fibroblast cells. Importantly, this pathway shows fast kinetics, is transcription-independent and is exclusively activated during ECM detachment, but not by canonical endoplasmic reticulum stressors. Moreover, enforced PERK or AMPK activation upregulates autophagy and causes luminal filling during acinar morphogenesis by perpetuating a population of surviving autophagic luminal cells that resist anoikis. Hence, we identify a novel pathway in which suspension-activated PERK promotes the activation of LKB1, AMPK and TSC2, leading to the rapid induction of detachment-induced autophagy. We propose that increased autophagy, secondary to persistent PERK and LKB1-AMPK signaling, can robustly protect cells from anoikis and promote luminal filling during early carcinoma progression.


Subject(s)
Autophagy , Extracellular Matrix/metabolism , Multiprotein Complexes/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , eIF-2 Kinase/metabolism , AMP-Activated Protein Kinases , Animals , Cell Adhesion , Cell Line, Tumor , Disease Progression , Female , Homeostasis , Humans , Lactation , Mammary Glands, Animal/embryology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/physiology , Mammary Neoplasms, Animal/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Organogenesis , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
6.
Cell Death Dis ; 3: e321, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22695614

ABSTRACT

Vascular deposition of amyloid-ß (Aß) in sporadic and familial Alzheimer's disease, through poorly understood molecular mechanisms, leads to focal ischemia, alterations in cerebral blood flow, and cerebral micro-/macro-hemorrhages, significantly contributing to cognitive impairment. Here, we show that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptors DR4 and DR5 specifically mediate oligomeric Aß induction of extrinsic apoptotic pathways in human microvascular cerebral endothelial cells with activation of both caspase-8 and caspase-9. The caspase-8 inhibitor cellular FLICE-like inhibitory protein (cFLIP) is downregulated, and mitochondrial paths are engaged through BH3-interacting domain death agonist (Bid) cleavage. Upregulation of DR4 and DR5 and colocalization with Aß at the cell membrane suggests their involvement as initiators of the apoptotic machinery. Direct binding assays using receptor chimeras confirm the specific interaction of oligomeric Aß with DR4 and DR5 whereas apoptosis protection achieved through RNA silencing of both receptors highlights their active role in downstream apoptotic pathways unveiling new targets for therapeutic intervention.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Endothelial Cells/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Alzheimer Disease/pathology , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Caspases/metabolism , Endothelial Cells/pathology , Humans , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor/genetics , Up-Regulation
7.
FASEB J ; 24(1): 229-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19770225

ABSTRACT

Cerebral amyloid angiopathy (CAA) is an age-associated condition and a common finding in Alzheimer's disease in which amyloid-beta (Abeta) vascular deposits are featured in >80% of the cases. Familial Abeta variants bearing substitutions at positions 21-23 are primarily associated with CAA, although they manifest with strikingly different clinical phenotypes: cerebral hemorrhage or dementia. The recently reported Piedmont L34V Abeta mutant, located outside the hot spot 21-23, shows a similar hemorrhagic phenotype, albeit less aggressive than the widely studied Dutch E22Q variant. We monitored the apoptotic events occurring after stimulation of human brain microvascular endothelial and smooth muscle cells with nonfibrillar structures of both variants and wild-type Abeta40. Induction of analogous caspase-mediated mitochondrial pathways was elicited by all peptides, although within different time frames and intensity. Activated pathways were susceptible to pharmacological modulation either through direct inhibition of mitochondrial cytochrome c release or by the action of pan- and pathway-specific caspase inhibitors, giving a clear indication of the independent or synergistic engagement of both extrinsic and intrinsic mechanisms. Structural analyses of the Abeta peptides showed that apoptosis preceded fibril formation, correlating with the presence of oligomers and/or protofibrils. The data support the notion that rare genetic mutations constitute unique paradigms to understand the molecular pathogenesis of CAA.


Subject(s)
Amyloid beta-Peptides/genetics , Brain/blood supply , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Apoptosis , Brain/metabolism , Brain/pathology , Caspases/metabolism , Cell Line , Cerebral Amyloid Angiopathy, Familial/metabolism , Cytochromes c/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Variation , Humans , Mitochondria/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 106(29): 12145-50, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19581601

ABSTRACT

A number of distinct beta-amyloid (Abeta) variants or multimers have been implicated in Alzheimer's disease (AD), and antibodies recognizing such peptides are in clinical trials. Humans have natural Abeta-specific antibodies, but their diversity, abundance, and function in the general population remain largely unknown. Here, we demonstrate with peptide microarrays the presence of natural antibodies against known toxic Abeta and amyloidogenic non-Abeta species in plasma samples and cerebrospinal fluid of AD patients and healthy controls aged 21-89 years. Antibody reactivity was most prominent against oligomeric assemblies of Abeta and pyroglutamate or oxidized residues, and IgGs specific for oligomeric preparations of Abeta1-42 in particular declined with age and advancing AD. Most individuals showed unexpected antibody reactivities against peptides unique to autosomal dominant forms of dementia (mutant Abeta, ABri, ADan) and IgGs isolated from plasma of AD patients or healthy controls protected primary neurons from Abeta toxicity. Aged vervets showed similar patterns of plasma IgG antibodies against amyloid peptides, and after immunization with Abeta the monkeys developed high titers not only against Abeta peptides but also against ABri and ADan peptides. Our findings support the concept of conformation-specific, cross-reactive antibodies that may protect against amyloidogenic toxic peptides. If a therapeutic benefit of Abeta antibodies can be confirmed in AD patients, stimulating the production of such neuroprotective antibodies or passively administering them to the elderly population may provide a preventive measure toward AD.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies/immunology , Neuroprotective Agents/immunology , Peptides/immunology , Aging/drug effects , Alzheimer Disease/blood , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Antibodies/blood , Antibodies/cerebrospinal fluid , Cytoprotection/drug effects , Dementia/complications , Dementia/immunology , Disease Progression , Genes, Dominant , Immunization , Immunoglobulin G/blood , Mice , Molecular Weight , Neurons/cytology , Neurons/drug effects , Peptides/chemistry , Primates/immunology , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary
9.
Cell Mol Life Sci ; 66(6): 1094-104, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19189048

ABSTRACT

The vasculotropic E22Q mutant of the amyloid-beta (Abeta) peptide is associated with hereditary cerebral hemorrhage with amyloidosis Dutch type. The cellular mechanism(s) of toxicity and nature of the AbetaE22Q toxic assemblies are not completely understood. Comparative assessment of structural parameters and cell death mechanisms elicited in primary human cerebral endothelial cells by AbetaE22Q and wild-type Abeta revealed that only AbetaE22Q triggered the Bax mitochondrial pathway of apoptosis. AbetaE22Q neither matched the fast oligomerization kinetics of Abeta42 nor reached its predominant beta-sheet structure, achieving a modest degree of oligomerization with a secondary structure that remained a mixture of beta and random conformations. The endogenous molecule tauroursodeoxycholic acid (TUDCA) was a strong modulator of AbetaE22Q-triggered apoptosis but did not significantly change the secondary structures and fibrillogenic propensities of Abeta peptides. These data dissociate the pro-apoptotic properties of Abeta peptides from their distinct mechanisms of aggregation/fibrillization in vitro, providing new perspectives for modulation of amyloid toxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/blood supply , Endothelial Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cerebellum/cytology , Cytochromes c/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Microvessels/cytology , Mitochondria/metabolism , Mutation , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Transport , bcl-2-Associated X Protein/metabolism
10.
Neuropathol Appl Neurobiol ; 34(5): 492-505, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18282158

ABSTRACT

INTRODUCTION: Two different disease-specific mutations in the BRI2 gene, situated on chromosome 13, have been identified as giving rise to familial British dementia (FBD) and familial Danish dementia (FDD). Each mutation results in extension of the open reading frame generating the disease-specific precursor proteins which are cleaved by furin-like proteolysis releasing the amyloidogenic C-terminal peptides ABri and ADan in FBD and FDD, respectively. MATERIAL AND METHODS: To understand the mechanism of the formation of amyloid lesions in FBD, we studied the origin of the precursor proteins and furin in the human brain. We used control brains, cases of sporadic Alzheimer's disease (AD), variant AD with cotton wool plaques and FBD to study BRI2 mRNA expression using in situ hybridization. Furin and BRI2 protein expression was investigated using Western blotting and immunohistochemistry. RESULTS: BRI2 mRNA and BRI2 protein are widely expressed primarily by neurones and glia and are deposited in the amyloid lesions in FBD. They were, however, not expressed by cerebrovascular components. Furin expression showed a similar pattern except that it was also present in cerebrovascular smooth muscle cells. CONCLUSIONS: These findings suggest that neurones and glia and are a major source of BRI2 protein and that in FBD, the mutated precursor protein may undergo furin cleavage within neurones to produce the amyloid peptide ABri. The failure to demonstrate BRI2 in blood vessels under the conditions tested suggests that vascular amyloid peptide production does not contribute significantly to cerebral amyloid angiopathy (CAA) in FBD and FDD, lending indirect support to the drainage hypothesis of CAA.


Subject(s)
Brain/metabolism , Dementia/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , RNA, Messenger/biosynthesis , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Blotting, Northern , Blotting, Western , Brain/pathology , Dementia/genetics , Dementia/pathology , Female , Fluorescent Antibody Technique , Furin/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Male , Membrane Glycoproteins , Middle Aged , Mutation , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Polymerase Chain Reaction , RNA, Messenger/analysis
11.
Neuropathol Appl Neurobiol ; 32(5): 492-504, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16972883

ABSTRACT

Molecular chaperons or amyloid-associated proteins (AAPs) are deposited in vascular and parenchymal amyloid lesions in Alzheimer's disease (AD) and other amyloidoses. AAPs, such as apolipoprotein E (ApoE) or apolipoprotein J (ApoJ) have been strongly implicated in the pathogenesis of AD in vitro and in vivo. Furthermore the possession of the ApoE in4 allele is a well-studied risk factor for AD. In view of the similarities between AD and both familial British dementia (FBD) and familial Danish dementia (FDD), we investigated the presence of AAPs in these two diseases to understand better their role in the general process of amyloidogenesis. Immunohistochemistry for ApoE, ApoJ, serum amyloid P (SAP), alpha-1-antichymotrypsin, cystatin C, heparan sulphate proteoglycans, such as agrin, perlecan, syndecans, glypican-1 and for heparan sulphate glycosaminoglycan (HS GAG) side chains was carried out together with immunohistochemical preparations specific to the amyloid subunits. Significant or extensive staining for ApoE, ApoJ, agrin, glypican-1 and HS GAG side chains was found in both amyloid (fibrillar) and preamyloid (nonfibrillar) deposits in FBD and FDD. The remaining AAPs, including SAP, were predominantly found in amyloid lesions. Only very weak staining was present in a small proportion of the amyloid lesions using perlecan immunohistochemistry. These findings suggest that the deposition patterns of AAPs in FBD and FDD are mostly similar to those in AD. The presence of AAPs in the preamyloid lesions supports the notion that chaperon molecules may play a role in the early steps of fibrillogenesis.


Subject(s)
Amyloid Neuropathies/pathology , Dementia/genetics , Dementia/pathology , Molecular Chaperones/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid/genetics , Amyloid/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Conformation , Proteoglycans/metabolism , Serum Amyloid P-Component/metabolism , Syndecans , alpha 1-Antichymotrypsin/metabolism
12.
Brain Pathol ; 16(1): 71-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16612984

ABSTRACT

Classic arguments sustaining the importance of amyloid in the pathogenesis of dementia are usually centered on amyloid beta (Abeta) and its role in neuronal loss characteristic of Alzheimer disease, the most common form of human cerebral amyloidosis. Two non-Abeta cerebral amyloidoses, familial British and Danish dementias, share many aspects of Alzheimer disease, including the presence of neurofibrillary tangles, parenchymal pre-amyloid and amyloid deposits, cerebral amyloid angiopathy, and a widespread inflammatory response. Both early-onset conditions are linked to specific mutations in the BRI2 gene, causing the generation of longer-than-normal protein products and the release of 2 de novo created peptides ABri and ADan, the main components of amyloid fibrils in these inherited dementias. Although the molecular mechanisms and signal transduction pathways elicited by the amyloid deposits and their relation to cognitive impairment remain to be clarified, new evidence indicates that, independent of the differences in their primary structures, Abeta, ABri, and ADan subunits are able to form morphologically compatible ion-channel-like structures and elicit single ion-channel currents in reconstituted lipid membranes. These findings reaffirm the notion that non-Abeta amyloidosis constitute suitable alternative models to study the role of amyloid deposition in the mechanism of neuronal cell death.


Subject(s)
Amyloid/genetics , Cerebral Amyloid Angiopathy/genetics , Dementia/genetics , Adaptor Proteins, Signal Transducing , Animals , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Dementia/pathology , Denmark , Humans , Membrane Glycoproteins , Membrane Proteins , United Kingdom
13.
Cell Mol Life Sci ; 62(16): 1814-25, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15968464

ABSTRACT

The importance of cerebral amyloid deposition in the mechanism of neurodegeneration is still debatable. Classic arguments are usually centered on amyloid beta(Abeta) and its role in the neuronal loss characteristic of Alzheimer's disease, the most common form of human cerebral amyloidosis. Two non-Abeta cerebral amyloidoses, familial British and Danish dementias (FBD and FDD), share many aspects of Alzheimer's disease, including the presence of neurofibrillary tangles, parenchymal preamyloid and amyloid deposits, cerebral amyloid angiopathy and a variety of amyloid-associated proteins and inflammatory components. Both early-onset conditions are linked to specific mutations at or near the stop codon of the chromosome 13 gene BRI2 that cause generation of longer-than-normal protein products. Furin-like processing of these longer precursors releases two de novo-created peptides, ABri and ADan, which deposit as amyloid fibrils in FBD and FDD, respectively. Due to the similar pathology generated by completely unrelated amyloid subunits, FBD and FDD, collectively referred to as chromosome 13 dementias, constitute alternative models for studying the role of amyloid deposition in the mechanism of neuronal cell death.


Subject(s)
Amyloid/metabolism , Chromosomes, Human, Pair 13/genetics , Dementia/genetics , Adaptor Proteins, Signal Transducing , Amyloid/genetics , Animals , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/pathology , Dementia/metabolism , Dementia/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Membrane Glycoproteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , Protein Subunits/genetics , Protein Subunits/metabolism
15.
Neurobiol Aging ; 23(3): 405-12, 2002.
Article in English | MEDLINE | ID: mdl-11959403

ABSTRACT

A point mutation of G to C at codon 693 of the amyloid-beta (Abeta) precursor protein gene results in Glu to Gln substitution at position 22 of the Abeta (AbetaQ22) associated with hereditary cerebrovascular amyloidosis with hemorrhage Dutch type. Factors that regulate AbetaQ22 levels in the central nervous system (CNS) are largely unknown. By using ventriculo-cisternal perfusion technique in guinea pigs, we demonstrated that clearance from the cerebrospinal fluid and transport from the CNS to blood of [(125)I]-AbetaQ22 (1 nM) were reduced by 36% and 52%, respectively, in comparison to the wild type Abeta(1-40) peptide. In contrast to significant uptake and transport of Abeta(1-40) across the brain capillaries and leptomeningeal vessels, AbetaQ22 was not taken up at these CNS vascular transport sites, which was associated with its 53% greater accumulation in the brain. The CNS clearance of Abeta(1-40) was half-saturated at 23.6 nM; AbetaQ22 had about 6.8-fold less affinity for the CNS efflux transporters and its elimination relied mainly on transport across the choroid plexus. Thus, the Dutch mutation impairs elimination of Abeta from brain by reducing its rapid transport across the blood-brain barrier and the vascular drainage pathways, which in turn may result in accumulation of the peptide around the blood vessels and in brain.


Subject(s)
Alzheimer Disease/genetics , Amino Acid Substitution/genetics , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Brain/metabolism , Cerebrovascular Circulation/genetics , Codon/genetics , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Amino Acid Sequence , Animals , Blood-Brain Barrier , Choroid Plexus/metabolism , Female , Guinea Pigs , Male , Molecular Sequence Data , Perfusion , Point Mutation , Protein Transport/genetics
16.
Biochemistry ; 40(49): 14995-5001, 2001 Dec 11.
Article in English | MEDLINE | ID: mdl-11732920

ABSTRACT

Inheritance of apoE4 is a strong risk factor for the development of late-onset sporadic Alzheimer's disease (AD). Several lines of evidence suggest that apoE4 binds to the Alzheimer Abeta protein and, under certain experimental conditions, promotes formation of beta-sheet structures and amyloid fibrils. Deposition of amyloid fibrils is a critical step in the development of AD. We report here that addition of melatonin to Abeta in the presence of apoE resulted in a potent isoform-specific inhibition of fibril formation, the extent of which was far greater than that of the inhibition produced by melatonin alone. This effect was structure-dependent and unrelated to the antioxidant properties of melatonin, since it could be reproduced neither with the structurally related indole N-acetyl-5-hydroxytryptamine nor with the antioxidants ascorbate, alpha-tocophenol, and PBN. The enhanced inhibitory effects of melatonin and apoE were lost when bovine serum albumin was substituted for apoE. In addition, Abeta in combination with apoE was highly neurotoxic (apoE4 > apoE3) to neuronal cells in culture, and this activity was also prevented by melatonin. These findings suggest that reductions in brain melatonin, which occur during aging, may contribute to a proamyloidogenic microenvironment in the aging brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Melatonin/pharmacology , Peptide Fragments/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Apolipoprotein E4 , Apolipoproteins E/genetics , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Circular Dichroism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Protein Structure, Secondary , Spectroscopy, Fourier Transform Infrared
17.
Amyloid ; 8 Suppl 1: 36-42, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11676288

ABSTRACT

The term cerebral amyloid angiopathy (CAA) refers to the specific deposition of amyloid fibrils in the walls of leptomeningeal and cortical arteries, arterioles and, although less frequently in capillaries and veins. It is commonly associated with Alzheimers disease, Down's syndrome and normal aging, as well as with a variety of familial conditions related to stroke and/or dementia: hereditary cerebral hemorrhage with amyloidosis of Icelandic type (HCHWA-I), various inherited disorders linked to Abeta mutants (including the Dutch variant of HCHWA), and the recently described chromosome 13 familial dementia in British and Danish kindreds. This review focuses on four different types of hereditary CAA, emphasizing the notion that CAA is not only related to stroke but also to neurodegeneration and dementia of the Alzheimer's type.


Subject(s)
Cerebral Amyloid Angiopathy, Familial/etiology , Dementia/etiology , Stroke/etiology , Alzheimer Disease/etiology , Amyloid beta-Peptides/genetics , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Cystatin C , Cystatins/genetics , Denmark , Genes, Dominant , Humans , Mutation , United Kingdom
18.
J Biol Chem ; 276(47): 43909-14, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11557758

ABSTRACT

Familial British dementia (FBD) is an early onset inherited disorder that, like familial Alzheimer's disease (FAD), is characterized by progressive dementia, amyloid deposition in the brain, and neurofibrillary degeneration of limbic neurons. The primary structure of the amyloid subunit (ABri) extracted from FBD brain tissues (Vidal, R., Frangione, B., Rostagno, A., Mead, S., Revesz, T., Plant, G., and Ghiso, J. (1999) Nature 399, 776-781) is entirely different and unrelated to any previously known amyloid protein. Patients with FBD have a single nucleotide substitution at codon 267 in the BRI2 gene, resulting in an arginine replacing the stop codon and a longer open reading frame of 277 amino acids instead of 266. The ABri peptide comprises the 34 C-terminal residues of the mutated precursor ABriPP-277 and is generated via furin-like proteolytic processing. Here we report that carriers of the Stop-to-Arg mutation have a soluble form of the amyloid peptide (sABri) in the circulation with an estimated concentration in the range of 20 ng/ml, several fold higher than that of soluble Abeta. In addition, ABri species identical to those identified in the brain were also found as fibrillar components of amyloid deposits predominantly in the blood vessels of several peripheral tissues, including pancreas and myocardium. We hypothesize that the high concentration of the soluble de novo created amyloidogenic peptide and/or the insufficient tissue clearance are the main causative factors for the formation of amyloid deposits outside the brain. Thus, FBD constitutes the first documented cerebral amyloidosis associated with neurodegeneration and dementia in which the amyloid deposition is also systemic.


Subject(s)
Amyloid/metabolism , Dementia/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Amyloid/genetics , Brain/pathology , Dementia/genetics , Dementia/pathology , Genetic Predisposition to Disease , Humans , Membrane Glycoproteins , Membrane Proteins , Molecular Sequence Data , Open Reading Frames , Peptide Fragments/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Neurochem ; 78(1): 121-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11432979

ABSTRACT

A large body of data suggests that the Alzheimer's amyloid peptide (Abeta) causes degeneration and death of neurons by mechanisms that involve reactive oxygen species. The pathways involved in Abeta-mediated oxidative injury are only partially understood. We theorized that abnormal microaggregates and/or pathological conformations of Abeta peptides may behave as xenobiotics and trigger the induction of NADPH cytochrome P450 reductase (CP450r), an enzyme which, if induced by non-physiological substrates (such as xenobiotics like drugs or other 'foreign molecules'), is known to cause oxidative stress. In order to test this hypothesis, i.e. that Abeta can increase the expression of CP450r, SK-N-SH human neuroblastoma cells were exposed to Abeta25-35 and Abeta1-42 and then examined for induction of this enzyme in immunoblots, using specific antibodies. Following exposure to Abeta peptides, neuroblastoma cells showed a clear-cut induction of CP450r. To determine whether this mechanism is operational in vivo, we investigated the expression of CP450r in a transgenic mouse model of Alzheimer's disease (AD) and in brains from patients afflicted with AD, using an immunocytochemical approach. Tissue sections from brains of transgenic mice exhibited strong immunoreactivity for CP450r, surrounding amyloid deposits. The pattern of expression of CP450r was similar to that exhibited by neuritic and oxidative stress markers. Sections from non-transgenic mice showed no detectable immunoreactivity. Immunostaining of sections from four brains with neuropathologically confirmed AD showed a pattern of abnormality different from transgenic mice that was characterized by abnormal immunoreactivity for CP450r within the cytoplasm of cortical neurons. No labeling was seen in sections from aged-matched control brains. The data showed that CP450r is induced by Alzheimer amyloid peptide and that such a response must be considered as one possible mechanism whereby Abeta causes oxidative stress.


Subject(s)
Amyloid beta-Peptides/pharmacology , NADPH-Ferrihemoprotein Reductase/metabolism , Peptide Fragments/pharmacology , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Brain/enzymology , Enzyme Induction , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic/genetics , Mutation/physiology , Peptide Fragments/genetics , Reference Values , Tumor Cells, Cultured
20.
Biochem Biophys Res Commun ; 283(4): 854-61, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11350063

ABSTRACT

Oncogenic transformation of fibroblasts by v-Src and v-Ras is often associated with downregulation of fibronectin (FN) and increased expression of CD44, a receptor for hyaluronan. Both v-Src and v-Ras as well as v-Raf activate phospholipase D through the small GTPase, RalA, an important mediator of transformation and tumorigenesis in vivo. We have therefore investigated whether RalA is involved in the downregulation of FN and overproduction of CD44 upon oncogenic transformation. We report here that compared to untransfected cells NIH3T3 cells transformed by v-Src, v-Ras, or v-Raf have reduced levels of FN and increased levels of CD44. Moreover, the ability to form extracellular FN fibrils was significantly reduced in the oncogene-transformed cells compared to parental controls. Coexpression of the dominant negative S28N-RalA mutant restored the levels of CD44 and FN and the capacity of v-Src-, v-Ras-, and v-Raf-expressing cells to form extracellular FN fibrils, to those observed in NIH3T3 cells. The data presented here show a novel regulatory role for RalA, which is required for tumor formation in transformed NIH3T3 cells, in mediating the signal transduction pathway activated by v-Src, v-Ras, and v-Raf, that leads to FN downregulation and CD44 overexpression.


Subject(s)
Fibronectins/metabolism , GTP Phosphohydrolases/physiology , Hyaluronan Receptors/metabolism , Oncogene Protein p21(ras)/metabolism , Oncogene Protein pp60(v-src)/metabolism , ral GTP-Binding Proteins , 3T3 Cells , Animals , Cell Line, Transformed , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...