Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 9(12): 1294-306, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12478466

ABSTRACT

The discovery of cell cycle regulators has directed cell research into uncharted territory. In dividing cells, cell cycle-associated protein kinases, which are referred to as cyclin-dependent-kinases (Cdks), regulate proliferation, differentiation, senescence and apoptosis. In contrast, all Cdks in post-mitotic neurons, with the notable exception of Cdk5, are silenced. Surprisingly, misregulation of Cdks occurs in neurons in a wide diversity of neurological disorders, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Ectopic expression of these proteins in neurons potently induces cell death with hallmarks of apoptosis. Deregulation of the unique, cell cycle-unrelated Cdk5 by its truncated co-activator, p25 and p29, contributes to neurodegeneration by altering the phosphorylation state of non-membrane-associated proteins and possibly through the induction of cell cycle proteins. On the other hand, cycling Cdks such as Cdk2, Cdk4 and Cdk6, initiate death pathways by derepressing E2F-1/Rb-dependent transcription at the neuronal G1/S checkpoint. Thus, Cdk5 and cycling Cdks may have little in common in the healthy CNS, but they likely conspire in leading neurons to their demise.


Subject(s)
Central Nervous System/metabolism , Cyclin-Dependent Kinases/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Central Nervous System/growth & development , Central Nervous System/physiopathology , Cyclin-Dependent Kinases/genetics , Genes, cdc/physiology , Humans , Neurodegenerative Diseases/genetics , Neurons/pathology , Signal Transduction/genetics
2.
J Cell Biol ; 155(2): 217-26, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11604419

ABSTRACT

Peripherin, a neuronal intermediate filament protein associated with axonal spheroids in amyotrophic lateral sclerosis (ALS), induces the selective degeneration of motor neurons when overexpressed in transgenic mice. To further clarify the selectivity and mechanism of peripherin-induced neuronal death, we analyzed the effects of peripherin overexpression in primary neuronal cultures. Peripherin overexpression led to the formation of cytoplasmic protein aggregates and caused the death not only of motor neurons, but also of dorsal root ganglion (DRG) neurons that were cultured from dissociated spinal cords of peripherin transgenic embryos. Apoptosis of DRG neurons containing peripherin aggregates was dependent on the proinflammatory central nervous system environment of spinal cultures, rich in activated microglia, and required TNF-alpha. This synergistic proapoptotic effect may contribute to neuronal selectivity in ALS.


Subject(s)
Apoptosis , Intermediate Filament Proteins/ultrastructure , Membrane Glycoproteins , Motor Neurons/ultrastructure , Nerve Tissue Proteins/ultrastructure , Tumor Necrosis Factor-alpha/physiology , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies/pharmacology , Cells, Cultured , Ganglia, Spinal/ultrastructure , Intermediate Filament Proteins/genetics , Mice , Mice, Transgenic , Microglia/metabolism , Microinjections , Nerve Tissue Proteins/genetics , Peripherins , Spinal Cord/physiology , Tumor Necrosis Factor-alpha/immunology
3.
J Neurochem ; 75(6): 2358-67, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080187

ABSTRACT

Previous studies have implicated stress-activated protein kinases (SAPKs) in aberrant phosphorylation of the high-molecular-mass neurofilament subunit (NFH). We now present direct evidence for this involvement using CEP-1347, a specific inhibitor of SAPK activation. Inhibition by this drug of stress-induced phosphorylation of NFH and the middle-molecular-mass neurofilament subunit in the perikaryon of dorsal root ganglion (DRG) neurons paralleled the decrease in levels of activated SAPKs and was essentially complete at 1 microM: CEP-1347. In addition, a role for SAPKs in the constitutive phosphorylation of NFH was demonstrated. Longterm treatment of unstressed DRG neurons with CEP-1347 lowered the steady-state phosphorylation level of NFH in neurites. No such effect was seen in neurons treated with PD 098059, which blocks activation of extracellular signal-regulated kinase 1/2. DRG neurites were shown to contain high basal levels of activated SAPKs. These included a 55-kDa SAPK whose activation was completely abolished at 0.05 microM: CEP-1347 and a 45-kDa SAPK that was not affected by the drug. These results indicate that SAPKs are involved in both stress-induced and constitutive phosphorylation of NFH. The differing responses of SAPKs in neurites and cell bodies to CEP-1347 inhibition further suggest the presence of different signaling pathways in the two neuronal compartments.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/drug effects , Indoles/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurofilament Proteins/metabolism , Animals , Axons/drug effects , Axons/metabolism , Cell Differentiation , Cells, Cultured , Fluorescent Antibody Technique , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Immunohistochemistry , Mitogen-Activated Protein Kinase 8 , PC12 Cells , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects
4.
J Neurochem ; 72(3): 1081-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10037479

ABSTRACT

PC12 cells are well characterized for their ability to differentiate into neuronal-like cells when challenged with nerve growth factor. It has been reported that the calpain and proteasome inhibitor N-acetyl-Leu-Leu-norleucinal (CI) is also able to induce neurite outgrowth in PC12 cells. In this study, we report that the inhibitor of proteasomal chymotrypsin-like activity, carbobenzoxy-Ile-Glu-(O-tert-butyl)-Ala-Leu-aldehyde (PSI), can also induce differentiation of PC12 cells. Induction of neurite outgrowth with PSI, CI, or its close analogue, carbobenzoxy-Leu-Leu-leucinal (MG132), was associated with stress-activated protein kinase (SAPK) activation. Neurite formation induced by protease inhibition was independent of mitogen-activated protein kinase/extracellular signal-regulated kinase, p38/reactivating kinase, or phosphatidylinositol 3-kinase activities. The exact mechanism by which protease inhibition activates SAPKs remains to be elucidated; however, our results suggest that the SAPK signal transduction cascade may be an alternative and/or parallel pathway in the regulation of neuronal differentiation.


Subject(s)
Mitogen-Activated Protein Kinases , Neurites/drug effects , Protease Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation/drug effects , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/physiology , Leupeptins/pharmacology , Mitogen-Activated Protein Kinase 12 , Neurites/enzymology , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Precipitin Tests , Protein Kinase Inhibitors , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
5.
J Neurochem ; 72(2): 693-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9930742

ABSTRACT

Mutations in the Cu/Zn-superoxide dismutase (SOD-1) gene underlie some familial cases of amyotrophic lateral sclerosis, a neurodegenerative disorder characterized by loss of cortical, brainstem, and spinal motor neurons. We present evidence that SOD-1 mutants alter the activity of molecular chaperones that aid in proper protein folding and targeting of abnormal proteins for degradation. In a cultured cell line (NIH 3T3), resistance to mutant SOD-1 toxicity correlated with increased overall chaperoning activity (measured by the ability of cytosolic extracts to prevent heat denaturation of catalase) as well as with up-regulation of individual chaperones/stress proteins. In transgenic mice expressing human SOD-1 with the G93A mutation, chaperoning activity was decreased in lumbar spinal cord but increased or unchanged in clinically unaffected tissues. Increasing the level of the stress-inducible chaperone 70-kDa heat shock protein by gene transfer reduced formation of mutant SOD-containing proteinaceous aggregates in cultured primary motor neurons expressing G93A SOD-1 and prolonged their survival. We propose that insufficiency of molecular chaperones may be directly involved in loss of motor neurons in this disease.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Chaperonins/metabolism , Superoxide Dismutase/genetics , 3T3 Cells , Animals , Cell Survival/physiology , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Humans , Mice , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/enzymology , Mutation/physiology , Neuroprotective Agents/metabolism , Spinal Cord/cytology , Superoxide Dismutase/metabolism , Transfection , Up-Regulation/physiology
6.
Article in English | MEDLINE | ID: mdl-9752717

ABSTRACT

This article reviews current knowledge of neurofilament structure, phosphorylation, and function and neurofilament involvement in disease. Neurofilaments are obligate heteropolymers requiring the NF-L subunit together with either the NF-M or the NF-H subunit for polymer formation. Neurofilaments are very dynamic structures; they contain phosphorylation sites for a large number of protein kinases, including protein kinase A (PKA), protein kinase C (PKC), cyclin-dependent kinase 5 (Cdk5), extracellular signal regulated kinase (ERK), glycogen synthase kinase-3 (GSK-3), and stress-activated protein kinase gamma (SAPK gamma). Most of the neurofilament phosphorylation sites, located in tail regions of NF-M and NF-H, consist of the repeat sequence motif, Lys-Ser-Pro (KSP). In addition to the well-established role of neurofilaments in the control of axon caliber, there is growing evidence based on transgenic mouse studies that neurofilaments can affect the dynamics and perhaps the function of other cytoskeletal elements, such as microtubules and actin filaments. Perturbations in phosphorylation or in metabolism of neurofilaments are frequently observed in neurodegenerative diseases. A down-regulation of mRNA encoding neurofilament proteins and the presence of neurofilament deposits are common features of human neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Parkinson's disease, and Alzheimer's disease. Although the extent to which neurofilament abnormalities contribute to pathogenesis in these human diseases remains unknown, emerging evidence, based primarily on transgenic mouse studies and on the discovery of deletion mutations in the NF-H gene of some ALS eases, suggests that disorganized neurofilaments can provoke selective degeneration and death of neurons. An interference of axonal transport by disorganized neurofilaments has been proposed as one possible mechanism of neurofilament-induced pathology. Other factors that can potentially lead to the accumulation of neurofilaments will be discussed as well as the emerging evidence for neurofilaments as being possible targets of oxidative damage by mutations in the superoxide dismutase enzyme (SOD1); such mutations are responsible for approximately 20% of familial ALS cases.


Subject(s)
Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Humans , Intermediate Filaments/genetics , Mice , Mice, Transgenic , Parkinson Disease/pathology , Sequence Deletion
7.
J Neurochem ; 70(5): 1869-75, 1998 May.
Article in English | MEDLINE | ID: mdl-9572270

ABSTRACT

We previously reported that activation of protein kinase A in cultured rat dorsal root ganglion neurons, treated concomitantly with low concentrations of okadaic acid that selectively inhibit protein phosphatase-2A, enhanced the Triton X-100 solubility of neurofilament triplet proteins. We now show that peripherin and alpha-internexin follow the same fragmentation profile as the neurofilament subunits, consistent with the notion that all five cytoplasmic intermediate filament proteins in these neurons form an integrated filamentous network whose assembly can be modulated by protein kinase A. Similar to the situation previously observed for the light neurofilament subunit, there was a strong correlation between phosphorylation of the amino-terminal head domain of peripherin and filament fragmentation. In contrast, insignificant levels of 32P were incorporated into alpha-internexin under conditions promoting disassembly, indicating that phosphorylation of this protein is not involved directly in filament fragmentation. The situation for the mid-sized neurofilament subunit (NFM) was not as clear-cut. Phosphopeptide mapping of NFM revealed many head and tail domain phosphorylation sites. However, changes in NFM head domain phosphorylation under conditions promoting filament disassembly were not as pronounced as for peripherin.


Subject(s)
Ganglia, Spinal/physiology , Intermediate Filament Proteins/physiology , Membrane Glycoproteins , Neurons/physiology , Animals , Carrier Proteins/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Molecular Weight , Nerve Tissue Proteins/physiology , Neurofilament Proteins/chemistry , Neurofilament Proteins/physiology , Okadaic Acid/pharmacology , Peptide Mapping , Peripherins , Phosphorylation , Rats
8.
Nucleic Acids Res ; 26(2): 486-9, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9421504

ABSTRACT

The immediate/early promoter/enhancer of cytomegalovirus (CMV promoter) is one of the most commonly used promoters for expression of transgenes in eukaryotic cells. In practice, the CMV promoter is often thought of as a constitutively active unregulated promoter. However, we have observed that transcription from the CMV promoter can be up-regulated by a variety of environmental stresses. Many forms of cellular stress stimulate MAP kinase signalling pathways, resulting in activation of stress-activated protein kinases [SAPKs, also called Jun N-terminal kinases (JNKs)] and p38 kinases. We have found that the same conditions that lead to activation of SAPK/JNKs and p38 kinases can also dramatically increase expression from the CMV promoter. Inhibitors of p38 kinases abolished basal transcription from the CMV promoter and completely blocked stress-induced up-regulation of the CMV promoter. Overexpression of a dominant negative JNK kinase had no effect on basal transcription, but significantly reduced up-regulation caused by stress. These results have grave implications for use of the CMV promoter. If the CMV promoter can be up-regulated by cellular stresses, inadvertent activation of the stress kinase pathways may complicate, if not invalidate, the interpretation of a wide range of experiments.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytomegalovirus/genetics , Gene Expression Regulation, Viral , Genes, Immediate-Early , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases , Promoter Regions, Genetic , 3T3 Cells , Animals , Cell Line , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Mice , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Transcription, Genetic , Transfection , beta-Galactosidase/genetics
9.
Neurosci Lett ; 229(2): 77-80, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9223595

ABSTRACT

The expression and Triton X-100 (Triton) solubility of neuronal intermediate filament proteins were determined in the developing rat cerebral cortex. The level of expression of alpha-internexin was unchanged from embryonic day 15 (E15) to postnatal day 15 (P15), whereas expression of the mid-sized neurofilament subunit increased continually during this interval concomitant with a reduction in Triton solubility of the two proteins. The low molecular weight neurofilament subunit, first barely detected at P2, was largely insoluble in Triton from the initial time point that its solubility could be assayed, at P5, to P15. Similar expression patterns and Triton solubility profiles were obtained for neuronal intermediate filament proteins in cultured neurons from E15 cerebral cortex. These results suggest that alpha-internexin is expressed earlier than neurofilament proteins to provide a more plastic network in the early developing brain. The incorporation of neurofilament proteins apparently results in the formation of the more stable intermediate filament network found in mature neurons.


Subject(s)
Carrier Proteins/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Neurofilament Proteins/metabolism , Age Factors , Animals , Intermediate Filament Proteins/metabolism , Rats , Rats, Sprague-Dawley
10.
J Neurosci Res ; 47(3): 300-10, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9039652

ABSTRACT

The developmental profile of the neurofilament (NF) triplet proteins, alpha-internexin and peripherin in cultured dorsal root ganglion neurons from gestation day 15 rat embryos was determined by Western blot analysis. At the outset (day 0 in culture), the neurons contained mostly alpha-internexin. A significant increase in peripherin levels was seen at days 1-2, in the midsized (NFM) and low molecular weight (NFL) NF subunits at days 2-3, and in the high molecular weight (NFH) NF subunit at days 5-6. Immunofluorescence microscopy showed that the five intermediate filament proteins were co-localized in all neuronal cell bodies and neurites. Analysis of Triton X-100 extracts from okadaic acid-treated dorsal root ganglion cultures revealed that peripherin and alpha-internexin followed the same fragmentation pattern observed with NFs. Interactions between the various neuronal intermediate filament proteins in these extracts were assessed by immunoprecipitation under native conditions using antibodies specific for the individual proteins. Co-immunoprecipitation of NFH with NFL, NFM with NFL, NFM with alpha-internexin, and alpha-internexin with peripherin demonstrated that the intermediate filament cytoskeleton in cultured sensory neurons is a highly integrated structure.


Subject(s)
Eye Proteins/metabolism , Intermediate Filament Proteins/metabolism , Membrane Glycoproteins , Nerve Tissue Proteins , Neurons/metabolism , Animals , Antibodies, Monoclonal , Antibody Specificity , Blotting, Western , Carrier Proteins/analysis , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cells, Cultured/chemistry , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Enzyme Inhibitors/pharmacology , Eye Proteins/analysis , Eye Proteins/immunology , Fluorescent Antibody Technique , Ganglia, Spinal/cytology , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/immunology , Neurofilament Proteins/analysis , Neurofilament Proteins/immunology , Neurofilament Proteins/metabolism , Neurons/chemistry , Neurons/cytology , Neurons/drug effects , Octoxynol , Okadaic Acid/pharmacology , Peripherins , Precipitin Tests , Rats
11.
J Neurobiol ; 32(2): 193-201, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9032661

ABSTRACT

The aim of this study was to assess the effects of low concentrations of okadaic acid (OA) on neurite outgrowth and cellular integrity in cultures of dissociated dorsal root ganglion (DRG) neurons. The complete and fully reversible arrest of neurite outgrowth was achieved at 1 nM OA, thus ruling out the involvement of protein phosphatase 1 in the observed inhibitory effect. OA at 0.5 nM did not completely block neurite outgrowth, although it reduced the rate of growth by about one third. Protein phosphorylation and the integrity of microtubules and neurofilaments in neuron-enriched cultures were unaffected by 1 nM OA. The rate of synthesis of the low-molecular-weight neurofilament subunit (NFL) was also unchanged by OA treatment. Antimitotic agents used to eliminate proliferating cells did not alter the rate of neurite elongation. Since 1 nM OA does not suffice to inhibit neuronal protein phosphatase 2A fully, owing to the high concentration of this enzyme in neurons, we propose that the inhibitor is affecting a neuronal compartment that contains low levels of the phosphatase. This putative compartment is likely to be located in neurites, which were shown to contain levels of protein phosphatase 2A that were two- to threefold lower than in neuronal perikarya.


Subject(s)
Cell Division/drug effects , Ganglia, Spinal/drug effects , Okadaic Acid/pharmacology , Animals , Cells, Cultured/drug effects , Rats , Rats, Sprague-Dawley
12.
J Neurosci ; 17(24): 9466-72, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9391002

ABSTRACT

The high-molecular-mass neurofilament subunit (NFH) is normally hypophosphorylated in the neuronal perikaryon and undergoes extensive phosphorylation after entering the initial axon segment. Aberrant hyperphosphorylation of perikaryal NFH is a common feature of many neurological diseases. In a previous study (), we demonstrated a correlation between phosphorylation of perikaryal NFH and induction of stress-activated protein kinase (SAPK)-gamma. In this report, we present direct evidence showing that the in vivo activation of SAPKs by an upstream activator (MEKK-1) caused extensive NFH phosphorylation. We also show that stress-activated p38 kinases were not involved in the phosphorylation of perikaryal NFH in cultured dorsal root ganglion neurons and that this process was reversible. SAPKgamma was shown to be located in both the cell body and the neurites of the cultured neurons, suggesting that it is likely to be involved in the phosphorylation of cytoplasmic substrates. These could include neuritic NFH, which is highly phosphorylated despite the demonstrated lack of cyclin-dependent kinase-5 activity in these neurons. Neuritic NFH was also highly phosphorylated in neuronal cultures devoid of Schwann cells, indicating that this form of post-translational modification does not require cues stemming from Schwann cell-axon contacts. Collectively, these findings provide significant new insights into mechanisms involved in NFH phosphorylation in normal neurons and in disease states characterized by aberrant phosphorylation of neurofilaments.


Subject(s)
Cyclin-Dependent Kinases , Mitogen-Activated Protein Kinases , Neurofilament Proteins/metabolism , Neurons/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3 Cells/physiology , Animals , Axons/chemistry , Axons/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 5 , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Mice , Mitogen-Activated Protein Kinase Kinases , Neurofilament Proteins/chemistry , Neurons/chemistry , Neurons/ultrastructure , Phosphorylation , Protein Structure, Tertiary , Rats , Stress, Physiological/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases
13.
J Biol Chem ; 272(49): 31073-8, 1997 Dec 05.
Article in English | MEDLINE | ID: mdl-9388258

ABSTRACT

Formation of protein dimers involving alpha-internexin, peripherin, and the neurofilament (NF) proteins NFH, NFM, and NFL was investigated by partial renaturation of various combinations of individually purified subunits in buffered 2 M urea. Oligomers that were formed were resolved by "blue" native electrophoresis (Schägger, H., Cramer, W. A., and von Jagow, G. (1994) Anal. Biochem. 217, 220-230) modified to include urea in the polyacrylamide gels. Combining this method with Western blot analysis, disulfide cross-linking, and SDS-polyacrylamide gel electrophoresis in the second dimension showed that NFL readily forms significant amounts of heterodimer with NFH, NFM, alpha-internexin, or peripherin in the presence of 2 M urea. alpha-Internexin and peripherin also formed heterodimers with NFH or NFM under these conditions. The modified version of blue native gel electrophoresis described here may be useful in monitoring the impact of post-translational modifications and mutations on the dimerization of intermediate filament proteins.


Subject(s)
Neurofilament Proteins/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Dimerization , Electrophoresis, Gel, Two-Dimensional , Intermediate Filament Proteins , Protein Binding , Protein Conformation , Rats
14.
J Biol Chem ; 271(48): 30404-9, 1996 Nov 29.
Article in English | MEDLINE | ID: mdl-8940004

ABSTRACT

The aberrant phosphorylation of the neurofilament high molecular weight subunit (NFH) in the neuronal perikaryon is a common feature of several neurological diseases. We demonstrated a strong correlation between hyperphosphorylation of the NFH carboxyl-terminal domain and activation of stress-activated protein kinase (SAPK) -gamma in PC12 cells. Agents that activated SAPKgamma in PC12 cells also caused the hyperphosphorylation of perikaryal NFH in cultured dorsal root ganglion neurons. The NFH carboxyl-terminal domain was phosphorylated by SAPKgamma in vitro, and the use of peptide substrates indicated that this event occurred preferentially at KSPXE motifs. We propose that SAPKgamma, perhaps in concert with other SAPKs, is involved in the abnormal phosphorylation of perikaryal NFH. This finding could lead to new insights into the etiology of several neurological diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Leupeptins/pharmacology , Mitogen-Activated Protein Kinases , Neurofilament Proteins/metabolism , Protein Kinases/metabolism , Stress, Physiological/metabolism , Amino Acid Sequence , Animals , Enzyme Activation/drug effects , Fluorescent Antibody Technique, Indirect , Ganglia, Spinal/metabolism , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 12 , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Neurons/metabolism , Neurons/ultrastructure , Oligopeptides/pharmacology , PC12 Cells , Phosphorylation , Protease Inhibitors/pharmacology , Rats , Structure-Activity Relationship
15.
J Neurochem ; 66(3): 1207-13, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8769885

ABSTRACT

The activation of cyclic AMP-dependent protein kinase (PKA) in rat dorsal root ganglion (DRG) cultures increased phosphorylation of the low-molecular-mass neurofilament subunit (NFL) at a site previously identified as Ser55 but had no effect on neurofilament integrity. When PKA was activated in DRG cultures treated with 20-250 nM okadaic acid, neurofilament fragmentation was enhanced, and there was a corresponding increase in phosphorylation of NFL at a novel site. This site was also phosphorylated by PKA in vitro and was determined to be Ser2 by mass spectrometric analysis of the purified chymotryptic phosphopeptide. The PKA sites in NFL were dephosphorylated by the purified catalytic subunit of protein phosphatase-2A but not that of protein phosphatase-1, and phosphoserine-2 was a better substrate than phosphoserine-55. The phosphorylation and dephosphorylation of Ser2 and Ser55 in NFL may therefore be involved in the modulation of neurofilament dynamics through the antagonistic effects of PKA and protein phosphatase-2A.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Okadaic Acid/pharmacology , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Enzyme Activation , Ganglia, Spinal/cytology , Neurofilament Proteins/chemistry , Neurons/drug effects , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Phosphatase 2 , Rats
16.
Int J Dev Neurosci ; 13(7): 753-8, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8787865

ABSTRACT

Treatment of PC12 cells or dorsal root ganglion neurons with the protease inhibitor, N-Acetyl-Leu-Leu-norleucinal, stimulated phosphorylation of the mid-sized and heavy neurofilament subunits and caused the heavy subunit in the perikarya of dorsal root ganglion neurons to become hyperphosphorylated. The closely related inhibitor, N-Acetyl-Leu-Leu-methioninal, did not produce a similar effect. Okadaic acid increased the phosphorylation state of the heavy neurofilament subunit in PC12 cells in a fashion similar to N-Acetyl-Leu-Leu-norleucinal and the effect of both compounds together was greater than for either one alone. There was no increase in cyclin-dependent kinase 5-immunoprecipitable histone H1 kinase activity in PC12 cells treated with N-Acetyl-Leu-Leu-norleucinal despite the presence of enzyme protein. The present study demonstrates that a protease inhibitor can induce the hyperphosphorylation of neurofilament subunits to a level normally seen only in axons. This suggests that perturbations in intracellular proteolysis may lead to the accumulation of phosphorylated neurofilament epitopes in neuronal perikarya in certain pathological states. The results also show that the carboxy-terminal tail domains of the two largest neurofilament subunits are phosphorylated even when cyclin dependent kinase 5 is inactive, indicating that other neuronal kinases are involved in the phosphorylation of Lys-Ser-Pro repeats.


Subject(s)
Ganglia, Spinal/metabolism , Leupeptins/pharmacology , Neurofilament Proteins/metabolism , Neurons/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Neurons/drug effects , Neurons/enzymology , PC12 Cells , Phosphorylation/drug effects , Precipitin Tests , Protein Kinases/metabolism , Rats , Stimulation, Chemical
17.
J Biol Chem ; 269(28): 18480-4, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8034596

ABSTRACT

Treatment of rat dorsal root ganglion cultures with 1 microM okadaic acid leads to a fragmentation of neurofilaments and a reduction in the electrophoretic mobilities of the three subunits on SDS-polyacrylamide gels (Sacher, M. G., Athlan, E. S., and Mushynski, W. E. (1992) Biochem. Biophys. Res. Commun. 186, 524-530). Based on the observed response to varying concentrations of okadaic acid, fragmentation was inferred to be due to inhibition of protein phosphatase-2A activity and reduction in electrophoretic mobility to inhibition of protein phosphatase-1. Okadaic acid treatment led to an increase in amino-terminal, relative to carboxyl-terminal, domain phosphorylation in the low molecular weight (NF-L) subunit in the Triton X-100-soluble and -insoluble fractions. The purified catalytic subunit of protein phosphatase-2A dephosphorylated 32P-labeled NF-L and the middle molecular weight subunit from okadaic acid-treated cultures, whereas the catalytic subunit of protein phosphatase-1 had no effect. In the case of NF-L, phosphate moieties were preferentially removed from the amino-terminal domain. These results show that the amino-terminal domain of NF-L can be phosphorylated in situ and implicate protein phosphatase-2A in the turnover of phosphate moieties in this domain.


Subject(s)
Ethers, Cyclic/toxicity , Neurofilament Proteins/metabolism , Neurons/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Animals , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Ganglia, Spinal/metabolism , Macromolecular Substances , Marine Toxins , Neurofilament Proteins/isolation & purification , Neurons/drug effects , Okadaic Acid , Oxazoles/pharmacology , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Phosphatase 2 , Rats
18.
Biochem Biophys Res Commun ; 186(1): 524-30, 1992 Jul 15.
Article in English | MEDLINE | ID: mdl-1321614

ABSTRACT

Treatment of 15-17 day old dissociated cultures of rat dorsal root ganglia with 1 microM okadaic acid caused a reduction in the mobilities of neurofilament subunits on SDS-polyacrylamide gels, signifying an increase in their phosphorylation levels. When cultures were exposed to okadaic acid for 0.5 hrs and harvested in buffer containing Triton X-100, NF-H was nearly completely redistributed to the detergent- soluble fraction while NF-M and NF-L required a longer exposure to the drug before undergoing a similar shift. This redistribution of subunits corresponded with striking changes in the immunofluorescence staining pattern for neurofilaments. Upon removal of okadaic acid from the culture medium following a 0.5 hr treatment, NF-L and NF-M returned to the Triton X-100 insoluble fraction within 2 hrs while NF-H required 10 hrs for recovery.


Subject(s)
Ethers, Cyclic/pharmacology , Ganglia, Spinal/ultrastructure , Intermediate Filaments/ultrastructure , Neurofilament Proteins/metabolism , Neurons/ultrastructure , Animals , Blotting, Western , Cells, Cultured , Detergents , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Embryo, Mammalian , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Intermediate Filaments/drug effects , Neurofilament Proteins/isolation & purification , Neurons/drug effects , Neurons/metabolism , Octoxynol , Okadaic Acid , Phosphoprotein Phosphatases/antagonists & inhibitors , Polyethylene Glycols , Rats
19.
Mol Cell Biol ; 12(3): 1239-47, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1545805

ABSTRACT

Translation initiation factor eIF-4E, which binds to the 5' cap structure of eukaryotic mRNAs, is believed to play an important role in the control of cell growth. Consistent with this, overexpression of eIF-4E in fibroblasts results in their malignant transformation. The activity of eIF-4E is thought to be regulated by phosphorylation on a single serine residue (Ser-53). Treatment of rat pheochromocytoma (PC12) cells with nerve growth factor (NGF) strongly curtails their growth and causes their differentiation into cells that resemble sympathetic neurons. The present study shows that eIF-4E is rapidly phosphorylated in PC12 cells upon NGF treatment, resulting in a significant increase in the steady-state levels of the phosphorylated protein. In contrast, epidermal growth factor, a factor which elicits a weak mitogenic response in PC12 cells, did not significantly enhance eIF-4E phosphorylation. We also show that although the mitogen and tumor promoter, phorbol 12-myristate-13-acetate, is able to induce phosphorylation of eIF-4E in PC12 cells, the NGF-mediated increase is primarily a protein kinase C-independent response. The NGF-induced enhancement of eIF-4E phosphorylation is abrogated in PC12 cells expressing a dominant inhibitory ras mutant (Ser-17 replaced by Asn), indicating that eIF-4E phosphorylation is dependent on a ras signalling pathway. As phosphorylation of eIF-4E effects translation initiation, these results suggest that NGF-mediated and ras-dependent eIF-4E phosphorylation may play a role in switching the pattern of gene expression during the differentiation of PC12 cells.


Subject(s)
Cell Differentiation , Genes, ras , Nerve Growth Factors/metabolism , Peptide Initiation Factors/metabolism , Animals , Eukaryotic Initiation Factor-4E , Isoelectric Focusing , Kinetics , PC12 Cells , Phosphorylation , Precipitin Tests , Protein Kinase C/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
20.
Acta Neuropathol ; 80(5): 499-505, 1990.
Article in English | MEDLINE | ID: mdl-2251907

ABSTRACT

Differential vulnerability of the major components of microtubules was examined in ischemic gerbil brains by a light microscopic, immunohistochemical method using monoclonal antibodies for microtubule-associated protein (MAP) 1A and MAP2, polyclonal antibody for MAP1 and 2 as well as monoclonal antibody for alpha-tubulin. Progressive cerebral ischemia during unilateral carotid occlusion for 5, 15 and 120 min and reperfusion for 3, 12 and 48 h following bilateral carotid occlusion for 10 min were studied. Ischemic lesions in the subiculum-CA1 region were visualized by all antibodies after ischemia for 5 min but the antibody for alpha-tubulin was less sensitive. The antibody for alpha-tubulin was also less sensitive than antibodies for MAPs for detection of early postischemic lesions. Differential sensitivity was also observed in the cerebral cortex and other brain regions. Microtubules in myelinated axons were more stable than those in dendrites. The observed loss of immunohistochemical reactivities for MAPs and alpha-tubulin may have been caused by activation of calcium-dependent proteolytic enzymes such as calpains. The discrepancy between MAPs and alpha-tubulin could be due to differences in affinities or topographic distributions of these proteins within microtubules.


Subject(s)
Brain Ischemia/metabolism , Microtubules/metabolism , Animals , Gerbillinae , Immunoenzyme Techniques , Microtubule-Associated Proteins/metabolism , Reperfusion , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...