Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 398
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Neuropathol Appl Neurobiol ; 44(5): 491-505, 2018 08.
Article in English | MEDLINE | ID: mdl-28755467

ABSTRACT

AIMS: Ageing-related tau astrogliopathy (ARTAG) appears in subependymal, subpial, perivascular, white matter (WM) and grey matter (GM) locations. Physical effects, blood-brain barrier dysfunction and blood- or vessel-related factors have been considered as aetiology. As connexin-43 (Cx43) and aquaporin-4 (AQP4) are related to these, we hypothesized that their immunoreactivity (IR) varies with ARTAG in a location-specific manner. METHODS: We performed a morphometric immunohistochemical study measuring the densities of IR of Cx43, AQP4, AT8 (phospho-tau) and glial fibrillar acidic protein (GFAP). We analysed the amygdala and hippocampus in age-matched cases with (n = 19) and without (n = 20) ARTAG in each of the locations it aggregates. RESULTS: We show a dramatic increase (>6-fold; P < 0.01) of Cx43 density of IR in ARTAG cases correlating strongly with AT8 density of IR, irrespective of the presence of neuronal tau pathology or reactive gliosis measured by GFAP density of IR, in the GM. In contrast, AQP4 density of IR was increased only in the WM and GM, and was associated with increased AT8 density of IR only in WM and perivascular areas. DISCUSSION: Our study reveals distinctive astroglial responses in each of the locations associated with ARTAG. Our observations support the concept that factors related to brain-fluid interfaces and water-ion imbalances most likely play a role in the generation of ARTAG. As Cx43 is crucial for maintaining neuronal homeostasis, the ARTAG-dependent increase of Cx43 density of IR suggests that the development of ARTAG in the GM most likely indicates an early response to the degeneration of neurons.


Subject(s)
Aging/pathology , Aquaporin 4/metabolism , Astrocytes/pathology , Brain/pathology , Connexin 43/metabolism , Tauopathies/pathology , Aged , Aged, 80 and over , Aging/metabolism , Aquaporin 4/analysis , Astrocytes/metabolism , Biomarkers/analysis , Brain/metabolism , Connexin 43/analysis , Female , Humans , Male , Tauopathies/metabolism
2.
Neuropathol Appl Neurobiol ; 43(4): 315-329, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27716988

ABSTRACT

AIMS: The aim of this study was to identify early foci of α-synuclein (α-syn pathology) accumulation, subsequent progression and neurodegeneration in multiple system atrophy of the cerebellar type (MSA-C). METHODS: We analysed 70-µm-thick sections of 10 cases with MSA-C and 24 normal controls. RESULTS: MSA-C cases with the lowest burden of pathology showed α-syn glial cytoplasmic inclusions (GCIs) in the cerebellum as well as in medullary and pontine cerebellar projections. Cerebellar pathology was highly selective and severely involved subcortical white matter, whereas deep white matter and granular layer were only mildly affected and the molecular layer was spared. Loss of Purkinje cells increased with disease duration and was associated with neuronal and axonal abnormalities. Neocortex, basal ganglia and spinal cord became consecutively involved with the increasing burden of α-syn pathology, followed by hippocampus, amygdala, and, finally, the visual cortex. GCIs were associated with myelinated axons, and the severity of GCIs correlated with demyelination. CONCLUSIONS: Our findings indicate that cerebellar subcortical white matter and cerebellar brainstem projections are likely the earliest foci of α-syn pathology in MSA-C, followed by involvement of more widespread regions of the central nervous system and neurodegeneration with disease progression.


Subject(s)
Cerebellum/pathology , Multiple System Atrophy/pathology , alpha-Synuclein , Aged , Central Nervous System/pathology , Disease Progression , Female , Humans , Male , Middle Aged , Nerve Degeneration/pathology
3.
Neuropathol Appl Neurobiol ; 43(7): 604-620, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28386933

ABSTRACT

AIMS: The aim of this study was to test the hypothesis that different conformations of misfolded α-synuclein (α-syn) are present in Parkinson's disease (PD) brain. METHODS: Using two previously characterized conformations of α-syn fibrils, we generated new conformation-selective α-syn monoclonal antibodies (mAbs). We then interrogated multiple brain regions in a well-characterized autopsy cohort of PD patients (n = 49) with these mAbs, Syn7015 and Syn9029. RESULTS: Syn7015 detects Lewy bodies (LBs) and Lewy neurites (LNs) formed by pathological α-syn in all brain regions tested, and is particularly sensitive to LNs and small Lewy dots, inclusions believed to form early in the disease. Further, we observed colocalization between Syn7015 and an early marker of α-syn pathology formation, phospho-Ser129-α-syn, and a lack of extensive colocalization with markers of more mature pathology. In comparison, Syn9029 detects Lewy pathology in all regions examined, but indicates significantly fewer LNs than Syn7015. In addition, colocalization of Syn9029 with later markers of α-syn pathology maturation (ubiquitin and P62) suggests that the pathology detected by Syn9029 is older. Semiquantitative scoring of both LN and LB pathology in nine brain regions further established this trend, with Syn7015 LN scores consistently higher than Syn9029 LN scores. CONCLUSIONS: Our data indicate that different conformations of α-syn pathology are present in PD brain and correspond to different stages of maturity for Lewy pathology. Regional analysis of Syn7015 and Syn9029 immunostaining also provides support for the Braak hypothesis that α-syn pathology advances through the brain.


Subject(s)
Lewy Bodies/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Brain/metabolism , Brain/pathology , Female , Humans , Lewy Bodies/metabolism , Male , Neurites/metabolism , Neurites/pathology , Primary Cell Culture , Protein Conformation , alpha-Synuclein/immunology
4.
Mol Psychiatry ; 20(8): 986-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869803

ABSTRACT

Synaptic roles for neurofilament (NF) proteins have rarely been considered. Here, we establish all four NF subunits as integral resident proteins of synapses. Compared with the population in axons, NF subunits isolated from synapses have distinctive stoichiometry and phosphorylation state, and respond differently to perturbations in vivo. Completely eliminating NF proteins from brain by genetically deleting three subunits (α-internexin, NFH and NFL) markedly depresses hippocampal long-term potentiation induction without detectably altering synapse morphology. Deletion of NFM in mice, but not the deletion of any other NF subunit, amplifies dopamine D1-receptor-mediated motor responses to cocaine while redistributing postsynaptic D1-receptors from endosomes to plasma membrane, consistent with a specific modulatory role of NFM in D1-receptor recycling. These results identify a distinct pool of synaptic NF subunits and establish their key role in neurotransmission in vivo, suggesting potential novel influences of NF proteins in psychiatric as well as neurological states.


Subject(s)
Brain/physiology , Motor Activity/physiology , Neurofilament Proteins/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Axons/drug effects , Axons/physiology , Brain/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Long-Term Potentiation/physiology , Mice, Knockout , Motor Activity/drug effects , Neurofilament Proteins/genetics , Receptors, Dopamine D1/metabolism , Synapses/drug effects , Synaptic Transmission/drug effects
5.
Dev Dyn ; 240(1): 52-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21117150

ABSTRACT

Sensory neurons transduce various stimuli including temperature, pain, and touch from the periphery to the central nervous system. Sensory neuron development is governed by a combination of extracellular cues and specific gene expression. We demonstrated that the transcription factor Sox11 was highly expressed in the developing sensory neurons. To test the function of Sox11, we used a knockin mouse model where the entire coding region of Sox11 was replaced by a LacZ reporter. The ablation of Sox11 caused severe reduction in sensory neuron survival in the trigeminal and dorsal root ganglia, although it did not affect migration of neural crest cells or acquisition of major sensory neuron subtypes. We further demonstrated that ablating Sox11 caused an arrest of axonal outgrowth in vivo and in vitro. This defect could not be fully rescued by blocking cell death. Our data suggest that Sox11 is a key regulator of sensory neuron development.


Subject(s)
Axons/physiology , Cell Proliferation , Nervous System/embryology , SOXC Transcription Factors/physiology , Sensory Receptor Cells/physiology , Animals , Axons/metabolism , Cell Death/genetics , Cell Movement/genetics , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Gene Knock-In Techniques , Mice , Mice, Knockout , Nervous System/growth & development , Neural Crest/cytology , Neural Crest/physiology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Sensory Receptor Cells/metabolism
6.
Neuropathol Appl Neurobiol ; 37(4): 358-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20942898

ABSTRACT

AIMS AND METHODS: The α-synucleinopathy multiple system atrophy (MSA) and diseases defined by pathological 43-kDa transactive response DNA-binding protein (TDP-43) or fused in sarcoma (FUS) aggregates such as amyotrophic lateral sclerosis and frontotemporal lobar degeneration show overlapping clinico-pathological features. Consequently, we examined MSA for evidence of TDP-43 or FUS pathology utilizing immunohistochemical studies in autopsy material from 29 MSA patients. RESULTS: TDP-43 pathology was generally rare, and there were no FUS lesions. The TDP-43 lesions were located predominantly in medio-temporal lobe and subcortical brain areas and were comprised mainly of dystrophic processes and perivascular (and subpial) lesions. CONCLUSIONS: The multisystem clinical symptoms and signs of MSA, and in particular the neurobehavioural/cognitive and pyramidal features, appear not to result from concomitant TDP-43 or FUS pathology, but rather from widespread white matter α-synuclein positive glial cytoplasmic inclusions and neurodegeneration in keeping with a primary α-synuclein-mediated oligodendrogliopathy. The gliodegenerative disease MSA evidently results from different pathogenetic mechanisms than neurodegenerative diseases linked to pathological TDP-43.


Subject(s)
Brain/pathology , Inclusion Bodies/pathology , Multiple System Atrophy/pathology , TDP-43 Proteinopathies/pathology , Aged , Brain/metabolism , DNA-Binding Proteins/metabolism , Female , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Male , Middle Aged , Multiple System Atrophy/complications , Multiple System Atrophy/metabolism , RNA-Binding Protein FUS/metabolism , TDP-43 Proteinopathies/complications , TDP-43 Proteinopathies/metabolism
8.
Nat Med ; 3(9): 1021-3, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9288730

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder involving the florid deposition of vascular and cerebral plaques composed chiefly of amyloid beta-peptide (A beta) derived from cleavage of the amyloid precursor protein (APP). Varying in length from 39 to 43 amino acids, A beta, particularly the longer A beta(42), is thought to play a significant role in AD pathogenesis. To better understand AD it is important to identify the subcellular organelles generating A beta. Studies using agents that disrupt endosomal/lysosomal function suggest that A beta is generated late in the secretory and endocytic pathways. However, much of what is known about A beta biosynthesis has been inferred by monitoring extracellular A beta levels since intracellular A beta is undetectable in most cell types. Consequently, the precise site or sites that generate A beta, or whether A beta(1-40) and A beta(1-42) are generated at the same point in the biosynthetic pathway, is not known. Using human NT2N neurons, we found that retention of APP in the endoplasmic reticulum/intermediate compartment (ER/IC) by three independent approaches eliminated production of intracellular A beta(1-40), but did not alter intracellular A beta(1-42) synthesis. These findings suggest that the ER/IC may be an important site for generating this highly amyloidogenic species of A beta.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Peptide Fragments/biosynthesis , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Base Sequence , Brefeldin A , Cell Compartmentation , Cell Line , Cyclopentanes/pharmacology , DNA Primers/genetics , Humans , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Neurons/drug effects , Neurons/ultrastructure , Peptide Fragments/genetics , Protein Synthesis Inhibitors/pharmacology
9.
J Cell Biol ; 141(4): 1031-9, 1998 May 18.
Article in English | MEDLINE | ID: mdl-9585420

ABSTRACT

The amyloid-beta peptide (Abeta) is produced at several sites within cultured human NT2N neurons with Abeta1-42 specifically generated in the endoplasmic reticulum/intermediate compartment. Since Abeta is found as insoluble deposits in senile plaques of the AD brain, and the Abeta peptide can polymerize into insoluble fibrils in vitro, we examined the possibility that Abeta1-40, and particularly the more highly amyloidogenic Abeta1-42, accumulate in an insoluble pool within NT2N neurons. Remarkably, we found that formic acid extraction of the NT2N cells solubilized a pool of previously undetectable Abeta that accounted for over half of the total intracellular Abeta. Abeta1-42 was more abundant than Abeta1-40 in this pool, and most of the insoluble Abeta1-42 was generated in the endoplasmic reticulum/intermediate compartment pathway. High levels of insoluble Abeta were also detected in several nonneuronal cell lines engineered to overexpress the amyloid-beta precursor protein. This insoluble intracellular pool of Abeta was exceptionally stable, and accumulated in NT2N neurons in a time-dependent manner, increasing 12-fold over a 7-wk period in culture. These novel findings suggest that Abeta amyloidogenesis may be initiated within living neurons rather than in the extracellular space. Thus, the data presented here require a reexamination of the prevailing view about the pathogenesis of Abeta deposition in the AD brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Animals , Brain/metabolism , CHO Cells , Carcinoma, Embryonal , Cell Line , Cricetinae , Endoplasmic Reticulum/metabolism , Genetic Vectors , Humans , Peptide Fragments/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Semliki forest virus , Solubility , Subcellular Fractions/metabolism , Transfection , Tumor Cells, Cultured
10.
J Cell Biol ; 139(5): 1307-15, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9382875

ABSTRACT

Mice engineered to express a transgene encoding a human Cu/Zn superoxide dismutase (SOD1) with a Gly93 --> Ala (G93A) mutation found in patients who succumb to familial amyotrophic lateral sclerosis (FALS) develop a rapidly progressive and fatal motor neuron disease (MND) similar to amyotrophic lateral sclerosis (ALS). Hallmark ALS lesions such as fragmentation of the Golgi apparatus and neurofilament (NF)-rich inclusions in surviving spinal cord motor neurons as well as the selective degeneration of this population of neurons were also observed in these animals. Since the mechanism whereby mutations in SOD1 lead to MND remains enigmatic, we asked whether NF inclusions in motor neurons compromise axonal transport during the onset and progression of MND in a line of mice that contained approximately 30% fewer copies of the transgene than the original G93A (Gurney et al., 1994). The onset of MND was delayed in these mice compared to the original G93A mice, but they developed the same neuropathologic abnormalities seen in the original G93A mice, albeit at a later time point with fewer vacuoles and more NF inclusions. Quantitative Western blot analyses showed a progressive decrease in the level of NF proteins in the L5 ventral roots of G93A mice and a concomitant reduction in axon caliber with the onset of motor weakness. By approximately 200 d, both fast and slow axonal transports were impaired in the ventral roots of these mice coincidental with the appearance of NF inclusions and vacuoles in the axons and perikarya of vulnerable motor neurons. This is the first demonstration of impaired axonal transport in a mouse model of ALS, and we infer that similar impairments occur in authentic ALS. Based on the temporal correlation of these impairments with the onset of motor weakness and the appearance of NF inclusions and vacuoles in vulnerable motor neurons, the latter lesions may be the proximal cause of motor neuron dysfunction and degeneration in the G93A mice and in FALS patients with SOD1 mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Axonal Transport/genetics , Intermediate Filaments/pathology , Mutation , Spinal Nerve Roots/pathology , Superoxide Dismutase/genetics , Age of Onset , Animals , Axons/pathology , Axons/physiology , Biological Transport , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gene Dosage , Humans , Mice , Mice, Transgenic , Motor Neuron Disease/etiology , Spinal Cord/pathology , Spinal Cord/physiology , Spinal Nerve Roots/physiology
11.
J Cell Biol ; 129(6): 1629-40, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7790359

ABSTRACT

Neurofilaments (NFs), the major intermediate filaments of central nervous system (CNS) and peripheral nervous system (PNS) neurons, are heteropolymers formed from the high (NFH), middle (NFM), and low (NFL) molecular weight NF subunits. To gain insights into how the expression of NF subunit proteins is regulated in vivo, two transgenes harboring coding sequences for human NFM (hNFM) with or without the hNFM multiphosphorylation repeat domain were introduced into mice. Expression of both hNFM constructs was driven by the hNFM promoter and resulted in increased levels of hNFM subunits concomitant with an elevation in the levels of mouse NFL (mNFL) proteins in the CNS of both lines of transgenic mice. The increased levels of mNFL appear specific to NFM because previous studies of transgenic mice overexpressing either NFL or NFH did not result in increased expression of either of the other two NF subunits. Further, levels of the most heavily phosphorylated isoforms of mouse NFH (mNFH) were reduced in the brains of these transgenic mice, and electron microscopic studies showed a higher packing density of NFs in large-diameter CNS axons of transgenic versus wild-type mice. Thus, reduced phosphorylation of the mNFH carboxy terminal domain may be a compensatory response of CNS neurons to the increase in NFs, and reduced negative charges on mNFH sidearms may allow axons to accommodate more NFs by increasing their packing density. Taken together, these studies imply that NFM may play a dominant role in the in vivo regulation of the levels of NFL protein, the stoichiometry of NF subunits, and the phosphorylation state of NFH. NFM and NFH proteins may assume similar functions in regulation of NF packing density in vivo.


Subject(s)
Brain/metabolism , Neurofilament Proteins/biosynthesis , Spinal Cord/metabolism , Aging/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Blotting, Northern , Blotting, Western , Brain/growth & development , Gene Expression , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Immunohistochemistry , Macromolecular Substances , Mice , Mice, Transgenic , Microscopy, Electron , Neurofilament Proteins/analysis , Neurofilament Proteins/chemistry , Organ Specificity , Phosphorylation , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Spinal Cord/growth & development
12.
J Cell Biol ; 138(3): 671-80, 1997 Aug 11.
Article in English | MEDLINE | ID: mdl-9245794

ABSTRACT

Previous studies have demonstrated that NT2N neurons derived from a human embryonal carcinoma cell line (NT2) constitutively process the endogenous wild-type beta-amyloid precursor protein (APP) to amyloid beta peptide in an intracellular compartment. These studies indicate that other proteolytic fragments generated by intracellular processing must also be present in these cells. Here we show that the NH2-terminal fragment of APP generated by beta-secretase cleavage (APPbeta) is indeed produced from the endogenous full length APP (APPFL). Pulse-chase studies demonstrated a precursor-product relationship between APPFL and APPbeta as well as intracellular and secreted APPbeta fragments. In addition, trypsin digestion of intact NT2N cells at 4 degrees C did not abolish APPbeta recovered from the cell lysates. Furthermore, the production of intracellular APPbeta from wild-type APP appears to be a unique characteristic of postmitotic neurons, since intracellular APPbeta was not detected in several non-neuronal cell lines. Significantly, production of APPbeta occurred even when APP was retained in the ER/ intermediate compartment by inhibition with brefeldin A, incubation at 15 degrees C, or by expression of exogenous APP bearing the dilysine ER retrieval motif.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Brefeldin A , CHO Cells , Cell Compartmentation , Cricetinae , Cyclopentanes/pharmacology , Glycosylation , Golgi Apparatus/metabolism , Humans , Neurons/enzymology , Protein Processing, Post-Translational , Tumor Cells, Cultured
13.
J Cell Biol ; 143(1): 195-205, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9763431

ABSTRACT

Neurofilaments (NFs) are prominent components of large myelinated axons. Previous studies have suggested that NF number as well as the phosphorylation state of the COOH-terminal tail of the heavy neurofilament (NF-H) subunit are major determinants of axonal caliber. We created NF-H knockout mice to assess the contribution of NF-H to the development of axon size as well as its effect on the amounts of low and mid-sized NF subunits (NF-L and NF-M respectively). Surprisingly, we found that NF-L levels were reduced only slightly whereas NF-M and tubulin proteins were unchanged in NF-H-null mice. However, the calibers of both large and small diameter myelinated axons were diminished in NF-H-null mice despite the fact that these mice showed only a slight decrease in NF density and that filaments in the mutant were most frequently spaced at the same interfilament distance found in control. Significantly, large diameter axons failed to develop in both the central and peripheral nervous systems. These results demonstrate directly that unlike losing the NF-L or NF-M subunits, loss of NF-H has only a slight effect on NF number in axons. Yet NF-H plays a major role in the development of large diameter axons.


Subject(s)
Axons/physiology , Axons/ultrastructure , Microtubules/physiology , Neurofilament Proteins/genetics , Neurofilament Proteins/physiology , Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Animals , Chimera , Exons , Genomic Library , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/ultrastructure , Neocortex/physiology , Neurofilament Proteins/deficiency , Restriction Mapping , Spinal Cord/physiology , Transfection
14.
J Cell Biol ; 141(3): 727-39, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9566972

ABSTRACT

Neurofilaments (NFs) are prominent components of large myelinated axons and probably the most abundant of neuronal intermediate filament proteins. Here we show that mice with a null mutation in the mid-sized NF (NF-M) subunit have dramatically decreased levels of light NF (NF-L) and increased levels of heavy NF (NF-H). The calibers of both large and small diameter axons in the central and peripheral nervous systems are diminished. Axons of mutant animals contain fewer neurofilaments and increased numbers of microtubules. Yet the mice lack any overt behavioral phenotype or gross structural defects in the nervous system. These studies suggest that the NF-M subunit is a major regulator of the level of NF-L and that its presence is required to achieve maximal axonal diameter in all size classes of myelinated axons.


Subject(s)
Axons/metabolism , Neurofilament Proteins/metabolism , Animals , Axons/ultrastructure , Cell Line , Gene Deletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neurofilament Proteins/genetics , Phenotype
15.
Science ; 251(4994): 675-8, 1991 Feb 08.
Article in English | MEDLINE | ID: mdl-1899488

ABSTRACT

Putative Alzheimer disease (AD)-specific proteins (A68) were purified to homogeneity and shown to be major subunits of one form of paired helical filaments (PHFs). The amino acid sequence and immunological data indicate that the backbone of A68 is indistinguishable from that of the protein tau (tau), but A68 could be distinguished from normal human tau by the degree to which A68 was phosphorylated and by the specific residues in A68 that served as phosphate acceptors. The larger apparent relative molecular mass (Mr) of A68, compared to normal human tau, was attributed to abnormal phosphorylation of A68 because enzymatic dephosphorylation of A68 reduced its Mr to close to that of normal tau. Moreover, the LysSerProVal motif in normal human tau appeared to be an abnormal phosphorylation site in A68 because the Ser in this motif was a phosphate acceptor site in A68, but not in normal human tau. Thus, the major subunits of a class of PHFs are A68 proteins and the excessive or inappropriate phosphorylation of normal tau may change its apparent Mr, thus transforming tau into A68.


Subject(s)
Alzheimer Disease/metabolism , Microtubule-Associated Proteins/metabolism , Amino Acid Sequence , Antibodies, Monoclonal , Blotting, Western , Humans , Intermediate Filaments/metabolism , Molecular Sequence Data , Molecular Weight , Phosphoproteins/metabolism , tau Proteins
16.
Science ; 290(5493): 985-9, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-11062131

ABSTRACT

Aggregated alpha-synuclein proteins form brain lesions that are hallmarks of neurodegenerative synucleinopathies, and oxidative stress has been implicated in the pathogenesis of some of these disorders. Using antibodies to specific nitrated tyrosine residues in alpha-synuclein, we demonstrate extensive and widespread accumulations of nitrated alpha-synuclein in the signature inclusions of Parkinson's disease, dementia with Lewy bodies, the Lewy body variant of Alzheimer's disease, and multiple system atrophy brains. We also show that nitrated alpha-synuclein is present in the major filamentous building blocks of these inclusions, as well as in the insoluble fractions of affected brain regions of synucleinopathies. The selective and specific nitration of alpha-synuclein in these disorders provides evidence to directly link oxidative and nitrative damage to the onset and progression of neurodegenerative synucleinopathies.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Antibodies, Monoclonal , Blotting, Western , Brain/pathology , Brain Chemistry , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Lewy Bodies/chemistry , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Microscopy, Immunoelectron , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/immunology , Neurons/chemistry , Neurons/metabolism , Neurons/ultrastructure , Parkinson Disease/metabolism , Parkinson Disease/pathology , Synucleins , Tyrosine/analysis , Tyrosine/immunology , alpha-Synuclein
17.
Science ; 282(5395): 1914-7, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9836646

ABSTRACT

Tau proteins aggregate as cytoplasmic inclusions in a number of neurodegenerative diseases, including Alzheimer's disease and hereditary frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). Over 10 exonic and intronic mutations in the tau gene have been identified in about 20 FTDP-17 families. Analyses of soluble and insoluble tau proteins from brains of FTDP-17 patients indicated that different pathogenic mutations differentially altered distinct biochemical properties and stoichiometry of brain tau isoforms. Functional assays of recombinant tau proteins with different FTDP-17 missense mutations implicated all but one of these mutations in disease pathogenesis by reducing the ability of tau to bind microtubules and promote microtubule assembly.


Subject(s)
Brain/metabolism , Dementia/genetics , Microtubules/metabolism , Parkinson Disease, Secondary/genetics , tau Proteins/genetics , tau Proteins/metabolism , Alternative Splicing , Cerebellum/metabolism , Chromosomes, Human, Pair 17 , Dementia/metabolism , Frontal Lobe/metabolism , Humans , Mutation , Mutation, Missense , Parkinson Disease, Secondary/metabolism , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Solubility , Syndrome , tau Proteins/chemistry
18.
Neuron ; 31(6): 885-8, 2001 Sep 27.
Article in English | MEDLINE | ID: mdl-11580890

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by the selective demise of specific neuronal populations leading to impairment of motor functions. Recent genetic studies have uncovered several genes involved in inherited forms of the disease. These gene products are implicated in the biochemical pathways underlying the etiology of sporadic PD. Mutations in the parkin gene causal of autosomal recessive juvenile parkinsonism highlight that ubiquitin-mediated proteolysis may play an important role in the pathobiology of PD.


Subject(s)
Cysteine Endopeptidases/metabolism , Ligases/physiology , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/physiology , Parkinson Disease/genetics , Ubiquitin-Protein Ligases , Age of Onset , Amino Acid Motifs , Animals , Chromosomes, Human, Pair 6/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genetic Predisposition to Disease , Humans , Lewy Bodies/metabolism , Ligases/deficiency , Ligases/genetics , Mice , Mice, Transgenic , Models, Biological , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Oxidative Stress , Parkinson Disease/epidemiology , Parkinson Disease/metabolism , Proteasome Endopeptidase Complex , Protein Folding , Rats , Structure-Activity Relationship , Substantia Nigra/metabolism , Synucleins , Ubiquitin/physiology
19.
Neuron ; 10(6): 1089-99, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8318230

ABSTRACT

Abnormally phosphorylated tau proteins (A68) are the building blocks of Alzheimer's disease (AD) paired helical filaments. The biological consequences of the conversion of normal adult tau to A68 remain unknown. Here we demonstrate that native A68 does not bind to microtubules (MTs), yet dephosphorylated A68 regains the ability to bind to MTs. Ser396 is phosphorylated in A68, but not in normal adult tau, whereas fetal tau is phosphorylated transiently at this site. Phosphorylation of tau at Ser396 by protein kinases in CHO cells and rat brain produces an electrophoretic mobility similar to that of A68. Using CHO cells transfected with an Ala396 mutant, we show that the phosphorylation of tau at Ser396 reduces its affinity for MTs and its ability to stabilize MTs against nocodazole-induced depolymerization. Our results demonstrate that the abnormal phosphorylation of tau in AD involves Ser396, and we suggest that this may be mediated by the inappropriate activation of fetal kinases or the reduced activity of tau protein phosphatases. Thus, phosphorylation of Ser396 may destabilize MTs in AD, resulting in the degeneration of affected cells.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Cerebral Cortex/metabolism , Microtubules/metabolism , Phosphoserine , Serine , tau Proteins/metabolism , Adult , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Fetus , Humans , Phosphorylation , Protein Binding , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , tau Proteins/biosynthesis , tau Proteins/isolation & purification
20.
Neuron ; 1(9): 817-25, 1988 Nov.
Article in English | MEDLINE | ID: mdl-2483104

ABSTRACT

Tau protein has been shown to be an integral component of Alzheimer paired helical filaments (PHF). However, the extent to which tau is incorporated into PHF has not been clear because the antibodies used to label PHF generally do not have precisely defined epitopes. Here we define the antigenic sites for five monoclonal antibodies that react with tau and cross-react with SDS-extracted neurofibrillary tangles. The reactive sites were determined by screening a lambda gt11 sublibrary expressing small fragments of the tau sequence. The mapped epitopes were found to span almost the entire length of tau, suggesting that PHF contains tau in its entirety or nearly in its entirety. One antibody was found to cross-react with microtubule-associated protein 2, implying some degree of homology between the two proteins.


Subject(s)
Alzheimer Disease/metabolism , Antibodies, Monoclonal , Epitopes , Microtubule-Associated Proteins/immunology , Neurofibrils/immunology , Amino Acid Sequence , Animals , Cattle , Humans , tau Proteins
SELECTION OF CITATIONS
SEARCH DETAIL