Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neuroscience ; 325: 30-8, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27012611

ABSTRACT

Brain tauopathies are characterized by abnormal processing of tau protein. While somatodendritic tau mislocalization has attracted considerable attention in tauopathies, the role of tau pathology in axonal transport, connectivity and related dysfunctions remains obscure. We have previously shown using the squid giant synapse that presynaptic microinjection of recombinant human tau protein (htau42) results in failure of synaptic transmission. Here, we evaluated molecular mechanisms mediating this effect. Thus, the initial event, observed after htau42 presynaptic injection, was an increase in transmitter release. This event was mediated by calcium release from intracellular stores and was followed by a reduction in evoked transmitter release. The effect of htau42 on synaptic transmission was recapitulated by a peptide comprising the phosphatase-activating domain of tau, suggesting activation of phosphotransferases. Accordingly, findings indicated that htau42-mediated toxicity involves the activities of both GSK3 and Cdk5 kinases.


Subject(s)
Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Synaptic Transmission/drug effects , tau Proteins/toxicity , Action Potentials/drug effects , Animals , Calcium/metabolism , Cyclin-Dependent Kinase 5/metabolism , Decapodiformes , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
2.
Neuroscience ; 284: 99-106, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25270904

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by progressive and irreversible cognitive and memory impairment. The discovery of familial forms of AD (fAD) in association with specific gene mutations facilitated the generation of numerous rodent models. These models in turn proved valuable for the study of molecular mechanisms underlying AD pathogenesis, and facilitated translational research and preclinical drug development. This study aimed to introduce a new rat model of AD simulating some aspects of the sporadic cases of disease. METHODS: Lentiviruses (LV) encoding human amyloid protein precursor (APP) bearing the fAD-linked Swedish and Indiana mutations (APPSw/Ind) were injected bilaterally in the hippocampus of adult rats. Passive avoidance and spatial memory performance were assessed 30 and 45 days post-injection, respectively. APP overexpression, intracellular accumulation of ß-amyloid (Aß) peptide, and astrogliosis were also evaluated using immunohistochemical procedures. RESULTS: Passive avoidance memory deficit was followed by impairments in spatial memory retrieval in LV (APPSw/Ind)-injected rats, compared to control animals. In addition, LV expression of APPSw/Ind was associated with intraneuronal accumulation of Aß, and reactive astrocytosis, two major AD hallmarks. CONCLUSION: Results from this work suggest that LV-mediated delivery of APPSw/Ind in adult rats represents a cost and time-effective animal model for the study of mechanisms underlying APP-linked fAD pathogenesis. The relevance of this animal model to the study of sporadic AD is discussed.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Disease Models, Animal , Mutation/genetics , Analysis of Variance , Animals , Avoidance Learning , Glial Fibrillary Acidic Protein/metabolism , Humans , Lentivirus/genetics , Male , Maze Learning/physiology , Rats , Rats, Transgenic , Rats, Wistar
3.
Oncogene ; 32(9): 1135-43, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-22525268

ABSTRACT

Neurofibromatosis type 2 patients develop schwannomas, meningiomas and ependymomas resulting from mutations in the tumor suppressor gene, NF2, encoding a membrane-cytoskeleton adapter protein called merlin. Merlin regulates contact inhibition of growth and controls the availability of growth factor receptors at the cell surface. We tested if microtubule-based vesicular trafficking might be a mechanism by which merlin acts. We found that schwannoma cells, containing merlin mutations and constitutive activation of the Rho/Rac family of GTPases, had decreased intracellular vesicular trafficking relative to normal human Schwann cells. In Nf2-/- mouse Schwann (SC4) cells, re-expression of merlin as well as inhibition of Rac or its effector kinases, MLK and p38(SAPK), each increased the velocity of Rab6 positive exocytic vesicles. Conversely, an activated Rac mutant decreased Rab6 vesicle velocity. Vesicle motility assays in isolated squid axoplasm further demonstrated that both mutant merlin and active Rac specifically reduce anterograde microtubule-based transport of vesicles dependent upon the activity of p38(SAPK) kinase. Taken together, our data suggest loss of merlin results in the Rac-dependent decrease of anterograde trafficking of exocytic vesicles, representing a possible mechanism controlling the concentration of growth factor receptors at the cell surface.


Subject(s)
Genes, Neurofibromatosis 2/physiology , MAP Kinase Kinase Kinases/metabolism , Microtubules/metabolism , Neurofibromin 2/genetics , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Decapodiformes , Humans , Mutation , Neurilemmoma/metabolism , Neurofibromin 2/metabolism
4.
Proc Natl Acad Sci U S A ; 106(14): 5907-12, 2009 Apr 07.
Article in English | MEDLINE | ID: mdl-19321417

ABSTRACT

The pathological mechanism by which Abeta causes neuronal dysfunction and death remains largely unknown. Deficiencies in fast axonal transport (FAT) were suggested to play a crucial role in neuronal dysfunction and loss for a diverse set of dying back neuropathologies including Alzheimer's disease (AD), but the molecular basis for pathological changes in FAT were undetermined. Recent findings indicate that soluble intracellular oligomeric Abeta (oAbeta) species may play a critical role in AD pathology. Real-time analysis of vesicle mobility in isolated axoplasms perfused with oAbeta showed bidirectional axonal transport inhibition as a consequence of endogenous casein kinase 2 (CK2) activation. Conversely, neither unaggregated amyloid beta nor fibrillar amyloid beta affected FAT. Inhibition of FAT by oAbeta was prevented by two specific pharmacological inhibitors of CK2, as well as by competition with a CK2 substrate peptide. Furthermore, perfusion of axoplasms with active CK2 mimics the inhibitory effects of oAbeta on FAT. Both oAbeta and CK2 treatment of axoplasm led to increased phosphorylation of kinesin-1 light chains and subsequent release of kinesin from its cargoes. Therefore pharmacological modulation of CK2 activity may represent a promising target for therapeutic intervention in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Axonal Transport/drug effects , Casein Kinase II/metabolism , Kinesins/metabolism , Neurons/pathology , Alzheimer Disease , Animals , Mice , Phosphorylation , Protein Multimerization
5.
Proc Natl Acad Sci U S A ; 104(7): 2442-7, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17287338

ABSTRACT

Parkinson's disease (PD), a late-onset condition characterized by dysfunction and loss of dopaminergic neurons in the substantia nigra, has both sporadic and neurotoxic forms. Neurotoxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and its metabolite 1-methyl-4-phenylpyridinium (MPP+) induce PD symptoms and recapitulate major pathological hallmarks of PD in human and animal models. Both sporadic and MPP+-induced forms of PD proceed through a "dying-back" pattern of neuronal degeneration in affected neurons, characterized by early loss of synaptic terminals and axonopathy. However, axonal and synaptic-specific effects of MPP+ are poorly understood. Using isolated squid axoplasm, we show that MPP+ produces significant alterations in fast axonal transport (FAT) through activation of a caspase and a previously undescribed protein kinase C (PKCdelta) isoform. Specifically, MPP+ increased cytoplasmic dynein-dependent retrograde FAT and reduced kinesin-1-mediated anterograde FAT. Significantly, MPP+ effects were independent of both nuclear activities and ATP production. Consistent with its effects on FAT, MPP+ injection in presynaptic domains led to a dramatic reduction in the number of membranous profiles. Changes in availability of synaptic and neurotrophin-signaling components represent axonal and synaptic-specific effects of MPP+ that would produce a dying-back pathology. Our results identify a critical neuronal process affected by MPP+ and suggest that alterations in vesicle trafficking represent a primary event in PD pathogenesis. We propose that PD and other neurodegenerative diseases exhibiting dying-back neuropathology represent a previously undescribed category of neurological diseases characterized by dysfunction of vesicle transport and associated with the loss of synaptic function.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Axonal Transport/drug effects , Caspases/metabolism , Protein Kinase C/metabolism , Animals , Decapodiformes , Disease Models, Animal , Enzyme Activation/drug effects , Kinetics , Neurons/pathology , Parkinson Disease/etiology , Parkinson Disease/pathology , Protein Kinase C-delta/metabolism , Synaptic Vesicles/drug effects
7.
Dev Neurosci ; 23(4-5): 364-76, 2001.
Article in English | MEDLINE | ID: mdl-11756752

ABSTRACT

Rapid organelle transport is required for process growth and establishment of specialized structures during neuronal development. Furthermore, maintenance of mature neuronal architecture and function depends on the proper delivery of materials to specialized domains within axons, such as nodes of Ranvier and synaptic terminals. Kinesin is the most abundant member of the kinesin superfamily of microtubule-based motors. Kinesins are responsible for anterograde transport of an assortment of membrane-bound organelles in all cell types. Kinesin is enriched in neurons, but relatively little is known about the developmental regulation of its expression, localization, and function in nervous tissue. By examining kinesin expression in developing brain and in cultures of cortical neurons, we found that kinesin is enriched in elongating neurites, including their growing tips, the growth cones. To gain understanding of mechanisms that underlie the delivery of proteins to specific cellular subcompartments, we focused on studying modifications on kinesin that lead to its dissociation from membranes. Since kinesin is a phosphoprotein in vivo, we evaluated the correlation between kinesin phosphorylation and its membrane association and identified a number of kinases which phosphorylate kinesin and alter its function.


Subject(s)
Aging/physiology , Cell Differentiation/physiology , Central Nervous System/embryology , Central Nervous System/growth & development , Growth Cones/metabolism , Intracellular Membranes/metabolism , Kinesins/metabolism , Animals , Animals, Newborn , Antibodies , Axonal Transport/physiology , Cell Movement/physiology , Central Nervous System/cytology , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/growth & development , Fetus , Growth Cones/ultrastructure , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Organelles/metabolism , Organelles/ultrastructure , Phosphorylation , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
8.
Mol Biol Cell ; 11(6): 2161-73, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848636

ABSTRACT

The nature of kinesin interactions with membrane-bound organelles and mechanisms for regulation of kinesin-based motility have both been surprisingly difficult to define. Most kinesin is recovered in supernatants with standard protocols for purification of motor proteins, but kinesin recovered on membrane-bound organelles is tightly bound. Partitioning of kinesin between vesicle and cytosolic fractions is highly sensitive to buffer composition. Addition of either N-ethylmaleimide or EDTA to homogenization buffers significantly increased the fraction of kinesin bound to organelles. Given that an antibody against kinesin light chain tandem repeats also releases kinesin from vesicles, these observations indicated that specific cytoplasmic factors may regulate kinesin release from membranes. Kinesin light tandem repeats contain DnaJ-like motifs, so the effects of hsp70 chaperones were evaluated. Hsc70 released kinesin from vesicles in an MgATP-dependent and N-ethylmaleimide-sensitive manner. Recombinant kinesin light chains inhibited kinesin release by hsc70 and stimulated the hsc70 ATPase. Hsc70 actions may provide a mechanism to regulate kinesin function by releasing kinesin from cargo in specific subcellular domains, thereby effecting delivery of axonally transported materials.


Subject(s)
Axonal Transport/physiology , Carrier Proteins/metabolism , HSP70 Heat-Shock Proteins , Kinesins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Detergents , Digitonin , Edetic Acid , Ethylmaleimide , Green Fluorescent Proteins , HSC70 Heat-Shock Proteins , Kinesins/isolation & purification , Luminescent Proteins/metabolism , Mice , Molecular Sequence Data , Octoxynol , Organelles/metabolism , Subcellular Fractions
9.
J Neurosci ; 18(23): 9858-69, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9822744

ABSTRACT

Cultures of cerebellar macroneurons were used to study the pattern of expression, subcellular localization, and function of the neuronal cdk5 activator p35 during laminin-enhanced axonal growth. The results obtained indicate that laminin, an extracellular matrix molecule capable of selectively stimulating axonal extension and promoting MAP1B phosphorylation at a proline-directed protein kinase epitope, selectively stimulates p35 expression, increases its association with the subcortical cytoskeleton, and accelerates its redistribution to the axonal growth cones. Besides, suppression of p35, but not of a highly related isoform designated as p39, by antisense oligonucleotide treatment selectively reduces cdk5 activity, laminin-enhanced axonal elongation, and MAP1b phosphorylation. Taken collectively, the present results suggest that cdk5/p35 may serve as an important regulatory linker between environmental signals (e.g., laminin) and constituents of the intracellular machinery (e.g., MAP1B) involved in axonal elongation.


Subject(s)
Axons/physiology , Cyclin-Dependent Kinases , Laminin/genetics , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Antibodies , Antisense Elements (Genetics) , Axons/chemistry , Cells, Cultured , Cerebellum/cytology , Cyclin-Dependent Kinase 5 , Epitopes/physiology , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/physiology , Laminin/analysis , Laminin/immunology , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neurons/ultrastructure , Phosphorylation , RNA, Messenger/analysis , Rabbits
10.
J Cell Biol ; 138(3): 657-69, 1997 Aug 11.
Article in English | MEDLINE | ID: mdl-9245793

ABSTRACT

In the present study, we present evidence about the cellular functions of KIF2, a kinesin-like superfamily member having a unique structure in that its motor domain is localized at the center of the molecule (Noda Y., Y. Sato-Yoshitake, S. Kondo, M. Nangaku, and N. Hirokawa. 1995. J. Cell Biol. 129:157-167.). Using subcellular fractionation techniques, isopicnic sucrose density centrifugation of microsomal fractions from developing rat cerebral cortex, and immunoisolation with KIF2 antibodies, we have now identified a type of nonsynaptic vesicle that associates with KIF2. This type of organelle lacks synaptic vesicle markers (synapsin, synaptophysin), amyloid precursor protein, GAP-43, or N-cadherin. On the other hand, it contains betagc, which is a novel variant of the beta subunit of the IGF-1 receptor, which is highly enriched in growth cone membranes. Both betagc and KIF2 are upregulated by NGF in PC12 cells and highly concentrated in growth cones of developing neurons. We have also analyzed the consequences of KIF2 suppression by antisense oligonucleotide treatment on nerve cell morphogenesis and the distribution of synaptic and nonsynaptic vesicle markers. KIF2 suppression results in a dramatic accumulation of betagc within the cell body and in its complete disappearance from growth cones; no alterations in the distribution of synapsin, synaptophysin, GAP-43, or amyloid percursor protein are detected in KIF2-suppressed neurons. Instead, all of them remained highly enriched at nerve terminals. KIF2 suppression also produces a dramatic inhibition of neurite outgrowth; this phenomenon occurs after betagc has disappeared from growth cones. Taken collectively, our results suggest an important role for KIF2 in neurite extension, a phenomenon that may be related with the anterograde transport of a type of nonsynaptic vesicle that contains as one of its components a growth cone membrane receptor for IGF-1, a growth factor implicated in nerve cell development.


Subject(s)
Kinesins/physiology , Neurites/physiology , Neurons/physiology , Organelles/metabolism , Amyloid beta-Protein Precursor/analysis , Animals , Antibodies, Monoclonal , Cerebral Cortex/chemistry , GAP-43 Protein , Kinesins/analysis , Kinesins/genetics , Kinesins/immunology , Membrane Glycoproteins/analysis , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/analysis , Neurons/cytology , Oligonucleotides, Antisense/pharmacology , Organelles/chemistry , PC12 Cells , Rats , Receptor, IGF Type 1/analysis , Subcellular Fractions/chemistry , Synapsins/analysis , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Synaptophysin/analysis , Thionucleotides/pharmacology , Up-Regulation
11.
J Neurosci Res ; 39(2): 219-32, 1994 Oct 01.
Article in English | MEDLINE | ID: mdl-7837290

ABSTRACT

Low density dissociated cultures of embryonic rat hippocampal cells were used to study the effects of neurotrophin-3 (NT-3) on neuronal morphogenesis. The results obtained indicate that NT-3 enhances neurite outgrowth and branching; this is a dose-dependent effect, detected in approximately 50% of the neurons, and prevented by K-252a, an inhibitor of the trk family of receptor protein kinases. NT-3 also accelerates the development of neuronal polarity, a phenomenon preceded by a dramatic accumulation of bundles of looped microtubules within axonal growth cones; these microtubule bundles contain tyrosinated, detyrosinated, and acetylated alpha-tubulin. Taken collectively, our data suggest that even though the basic shape of hippocampal neurons may be endogenously determined, critical aspects of their morphological development may be modulated by trophic factors such as NT-3. In addition, our observations suggest that at least some of the neuritogenic effects of NT-3 involve a stimulation of microtubule assembly and/or transport.


Subject(s)
Hippocampus/drug effects , Nerve Growth Factors/pharmacology , Neurites/drug effects , Pyramidal Cells/drug effects , Acetylation , Animals , Axons/drug effects , Axons/ultrastructure , CHO Cells , Carbazoles/pharmacology , Cell Polarity/drug effects , Cell Survival/drug effects , Cells, Cultured , Ciliary Neurotrophic Factor , Cricetinae , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/embryology , Indole Alkaloids , Microtubules/drug effects , Microtubules/ultrastructure , Morphogenesis/drug effects , Nerve Tissue Proteins/pharmacology , Neurotrophin 3 , Protein Processing, Post-Translational/drug effects , Pyramidal Cells/ultrastructure , Rats , Recombinant Proteins/pharmacology , Stimulation, Chemical , Tubulin/metabolism , Tyrosine/metabolism
12.
Cell Motil Cytoskeleton ; 29(2): 117-30, 1994.
Article in English | MEDLINE | ID: mdl-7820862

ABSTRACT

We report here a novel intracellular localization and function of Tau proteins in cultured cerebellar neurons. Immunofluorescence staining of detergent-extracted cytoskeletons with antibodies specific for Tau proteins revealed intense labeling of growth cone microtubules. Besides, suppression of Tau by antisense oligonucleotide treatment results in the complete disappearance of antigen 13H9, a specific growth cone component with properties of microfilament- and microtubule-associated protein [Goslin et al., 1989: J. Cell Biol. 109:1621-1631], from its normal intracellular location. This phenomenon is unique to neurite-bearing cells, is not associated with the disappearance of microtubules from growth cones, and is not reversed by taxol, a microtubule-stabilizing agent. In addition, Tau-suppressed neurons display a significant reduction in growth cone area and fillopodial number; on the contrary, fillopodial length increases significantly. The alterations in growth cone morphology are accompanied by considerable changes in the phalloidin staining of assembled actin. Taken together, the present results suggest that in developing neurons Tau proteins participate in mediating interactions between elements of the growth cone cytoskeleton important for maintaining the normal structural organization of this neuritic domain.


Subject(s)
Actin Cytoskeleton/physiology , Neurons/physiology , tau Proteins/physiology , Actin Cytoskeleton/chemistry , Actins/analysis , Animals , Antigens/analysis , Antigens/physiology , Cell Division/physiology , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/physiology , Cerebellar Cortex/ultrastructure , Cytoskeleton/chemistry , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/physiology , Microtubules/chemistry , Microtubules/physiology , Microtubules/ultrastructure , Neurons/chemistry , Neurons/cytology , Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , Paclitaxel/pharmacology , Rats , tau Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...