Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 20(1): 196-204, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10594022

ABSTRACT

We have previously shown that nerve growth factor (NGF) withdrawal-induced death requires the activity of the small GTP-binding protein Cdc42 and that overexpression of an active form of Cdc42 is sufficient to mediate neuronal apoptosis via activation of the c-Jun pathway. Recently, a new mitogen-activated protein (MAP) kinase kinase kinase, apoptosis signal-regulating kinase 1 (ASK1) which activates both the c-Jun N-terminal kinase (JNK) and p38 MAP kinase pathways and plays pivotal roles in tumor necrosis factor- and Fas-induced apoptosis, has been identified. Therefore, we investigated the role of ASK1 in neuronal apoptosis by using rat pheochromocytoma (PC12) neuronal cells and primary rat sympathetic neurons (SCGs). Overexpression of ASK1-DeltaN, a constitutively active mutant of ASK1, activated JNK and induced apoptosis in differentiated PC12 cells and SCG neurons. Moreover, in differentiated PC12 cells, NGF withdrawal induced a four- to fivefold increase in the activity of endogenous ASK1. Finally, expression of a kinase-inactive ASK1 significantly blocked both NGF withdrawal- and Cdc42-induced death and activation of c-jun. Taken together, these results demonstrate that ASK1 is a crucial element of NGF withdrawal-induced activation of the Cdc42-c-Jun pathway and neuronal apoptosis.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/pathology , Signal Transduction , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Animals , Cell Differentiation , Cells, Cultured , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinase 5 , MAP Kinase Signaling System , Neurons/metabolism , Rats , Rats, Sprague-Dawley , cdc42 GTP-Binding Protein/metabolism
2.
J Neuroimmunol ; 97(1-2): 119-28, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10408965

ABSTRACT

We examined the treatment effects of two structurally distinct phosphodiesterase type IV (PDE IV) inhibitors, BBB022 and rolipram, in murine and rat models of experimental autoimmune encephalomyelitis (EAE). Based on our data, we propose a mechanism of action which may supplement immunomodulatory effects of PDE IV inhibitors. In particular, PDE inhibitors promote elevation of intracellular cAMP levels, increasing the electrical resistance of endothelial monolayers by stabilizing intercellular junctional complexes. Such an effect on central nervous system (CNS) vascular endothelium has the potential to reduce disease severity in EAE, because both inflammatory cells and humoral factors readily cross a disrupted blood-brain barrier (BBB). In this report, we demonstrate the capacity of BBB022 and rolipram to decrease clinical severity of EAE. further, PDE IV inhibitors significantly reduced BBB permeability in the spinal cords of mice with EAE. These results provide evidence that PDE IV-inhibitors may exert therapeutic effects in EAE by modifying cerebrovascular endothelial permeability, reducing tissue edema as well as entry of inflammatory cells and factors.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Blood-Brain Barrier/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Acute Disease , Animals , Blood-Brain Barrier/immunology , Brain Edema/drug therapy , Brain Edema/immunology , Central Nervous System/enzymology , Central Nervous System/immunology , Chronic Disease , Cyclic Nucleotide Phosphodiesterases, Type 4 , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred Strains , Organic Chemicals , Rats , Rats, Inbred Lew , Recurrence , Rolipram
3.
Annu Rev Neurosci ; 22: 11-28, 1999.
Article in English | MEDLINE | ID: mdl-10202530

ABSTRACT

The blood-brain barrier (BBB) is formed by brain capillary endothelial cells (ECs). In the late embryonic and early postnatal period, these cells respond to inducing factors found in the brain environment by adopting a set of defined characteristics, including high-electrical-resistance tight junctions. Although the factors have not been identified definitively, a great deal of information about brain ECs has been obtained, especially recently. This review concentrates on a cell biological analysis of the BBB, with an emphasis on regulation of the specialized intercellular junctions. The development of these junctions seems to depend on two primary processes: the appearance of high levels of the tight junction protein occludin and intracellular signaling processes that control the state of phosphorylation of junctional proteins. Recent studies have revealed that the BBB can be modulated in an ongoing way to respond to environmental stimuli.


Subject(s)
Blood-Brain Barrier/physiology , Brain/blood supply , Capillaries/physiology , Endothelium, Vascular/physiology , Animals , Capillaries/cytology , Capillary Permeability/physiology , Endothelium, Vascular/cytology , Humans , Signal Transduction/physiology , Tight Junctions/physiology
4.
J Cell Biol ; 142(6): 1583-93, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9744886

ABSTRACT

Cytochrome c has been shown to play a role in cell-free models of apoptosis. During NGF withdrawal-induced apoptosis of intact rat superior cervical ganglion (SCG) neurons, we observe the redistribution of cytochrome c from the mitochondria to the cytoplasm. This redistribution is not inhibited by the caspase inhibitor Z-Val-Ala-Asp-fluoromethylketone (ZVADfmk) but is blocked by either of the neuronal survival agents 8-(4-chlorophenylthio)adenosine 3':5'-cyclic monophosphate (CPT-cAMP) or cycloheximide. Moreover, microinjection of SCG neurons with antibody to cytochrome c blocks NGF withdrawal-induced apoptosis. However, microinjection of SCG neurons with cytochrome c does not alter the rate of apoptosis in either the presence or absence of NGF. These data suggest that cytochrome c is an intrinsic but not limiting component of the neuronal apoptotic pathway.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Neurons/cytology , Adenosine Triphosphate/analogs & derivatives , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Biological Transport , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/antagonists & inhibitors , Cytoplasm/metabolism , Humans , Intracellular Membranes/physiology , Jurkat Cells , Membrane Potentials , Microinjections , Mitochondria/metabolism , Nerve Growth Factors/metabolism , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/pharmacology , Protein Synthesis Inhibitors/pharmacology , Rats , Superior Cervical Ganglion/cytology , Thionucleotides/pharmacology
5.
Am J Physiol ; 274(6): H1885-94, 1998 06.
Article in English | MEDLINE | ID: mdl-9841516

ABSTRACT

We tested the hypothesis that increased endothelial cell adenosine 3',5'-cyclic monophosphate (cAMP) decreases microvascular permeability in vivo. The effects of cAMP-specific phosphodiesterase type IV inhibition and adenylate cyclase activation on microvascular hydraulic conductivity (Lp) were investigated in intact individual capillaries and postcapillary venules in mesentery of pithed frogs (Rana pipiens). Treatment with rolipram (10 microM) and forskolin (5 microM) for 25 min decreased Lp to 37% of control. Rolipram alone also significantly decreased Lp. Isoproterenol (10 microM) decreased Lp to 27% of control within 20 min. A subgroup of eight vessels treated with rolipram and forskolin, in which mean Lp fell to 25% of control, was examined with transmission electron microscopy. The mean number of tight junctions in the treated vessels was 2.2 per cleft (303 clefts), significantly higher than in a matched control group (192 clefts), which was 1.7 per cleft. The results indicate that microvascular Lp can be modulated by intracellular cAMP and that one of the structural end points of stimulated cAMP levels is an increase in the mean number of tight-junction strands between endothelial cells.


Subject(s)
Capillary Permeability/physiology , Cyclic AMP/physiology , Lymphatic System/physiology , Tight Junctions/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Capillary Permeability/drug effects , Colforsin/pharmacology , Hemorheology , Isoproterenol/pharmacology , Lymphatic System/drug effects , Male , Pyrrolidinones/pharmacology , Rana pipiens , Rolipram , Tight Junctions/drug effects
6.
Proc Natl Acad Sci U S A ; 95(7): 3984-9, 1998 Mar 31.
Article in English | MEDLINE | ID: mdl-9520479

ABSTRACT

An increase in the level of the c-Jun transcription factor and of its phosphorylation has previously been shown to be essential for nerve growth factor (NGF) withdrawal-induced apoptosis of rat sympathetic neurons (SCG). The Rho-like GTPases Cdc42 and Rac1 are involved in the regulation of a number of cellular processes, including activation of the c-Jun NH2-terminal kinase (JNK) pathway. Therefore, we have investigated the role of these GTPases in this process. Overexpression of activated Rac1 or Cdc42 in SCG neurons maintained in the presence of NGF induced apoptosis, whereas expression of dominant negative mutants of Cdc42 or Rac1 blocked apoptosis following NGF withdrawal. Cdc42 activation produced an increase in the level of c-Jun and of its phosphorylation. Furthermore, Cdc42-induced death was prevented by coexpressing the c-Jun dominant negative FLAGDelta169. Thus, Cdc42 appears to function as an initiator of neuronal cell death by activating a transcriptional pathway regulated by c-Jun.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , GTP-Binding Proteins/metabolism , Ganglia/pathology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Nerve Growth Factors/metabolism , Neurons/pathology , Signal Transduction , Animals , Apoptosis/drug effects , Ganglia/metabolism , MAP Kinase Kinase 4 , Nerve Growth Factors/pharmacology , Neurons/metabolism , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , cdc42 GTP-Binding Protein
7.
Brain Res ; 787(2): 277-85, 1998 Mar 23.
Article in English | MEDLINE | ID: mdl-9518648

ABSTRACT

We examined the effect of BBB022, a type IV phosphodiesterase inhibitor, on blood-brain barrier (BBB) integrity after transient middle cerebral artery occlusion (MCAo) in rats. Male Sprague-Dawley rats were anesthetized with halothane and subjected to 120 min of temporary MCAo by retrograde intraluminal insertion of a nylon suture coated with poly-L-lysine. The drug (BBB022 in saline, 1 mg kg-1 h-1, i.v.) or vehicle (0.9% saline, 1-2 ml kg-1 h-1) was administered by infusion after the onset of MCAo. Four animal groups were studied: Groups A and B were treated by infusion of vehicle or drug over 5 h, and groups C and D over 48 h. Damage to the BBB was judged by extravasation of Evans blue (EB) dye, which was administered i.v. at 3 h after the onset of MCAo in groups A and B; and at 46 h in groups C and D. Fluorometric quantitation of EB was performed 1 or 2 h later in six brain regions. In the 5-h infusion series (group B), BBB022 decreased dye extravasation in the ipsilateral cortex, striatum and hemisphere (hemisphere mean+/-S.E.M. : 41.2+/-5.4 vs. 82.4+/-9.2 microg/g, p=0.005) compared to the vehicle-treated group (A). The 48-h infusion of BBB022 (group D) also decreased dye extravasation in the ipsilateral cortex (7.4+/-2. 5 vs. 29.0+/-8.3 microg/g, p=0.05), striatum (17.2+/-2.2 vs. 50. 8+/-12.1 microg/g, p=0.03) and hemisphere (30.7+/-4.0 vs. 93.2+/-18 microg/g, p=0.01) compared to the vehicle-treated group (C). BBB022 also significantly improved the neurological score at 3 and 5 h after MCAo (in the 5-h infusion group) and at 60 min, 24 h and 48 h (in the 48-h infusion group) compared to the vehicle groups. These data indicate that BBB022 prevents ischemic damage to the BBB after focal cerebral ischemia in rats.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Brain/pathology , Phosphodiesterase Inhibitors/therapeutic use , Pyrrolidinones/therapeutic use , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Animals , Coloring Agents , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Evans Blue , Male , Neostriatum/metabolism , Neostriatum/pathology , Organic Chemicals , Rats , Rats, Sprague-Dawley , Rolipram , Tight Junctions/drug effects , Tight Junctions/physiology
8.
J Neurosci ; 18(5): 1713-24, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9464996

ABSTRACT

When deprived of nerve growth factor (NGF), developing sympathetic neurons die by apoptosis. This death is associated with an increase in the level of c-Jun protein and is blocked by expression of a c-Jun dominant negative mutant. Here we have investigated whether NGF withdrawal activates Jun kinases, a family of stress-activated protein kinases that can stimulate the transcriptional activity of c-Jun by phosphorylating serines 63 and 73 in the transactivation domain and which can activate c-jun gene expression. We found that sympathetic neurons contained high basal levels of Jun kinase activity that increased further after NGF deprivation. In contrast, p38 kinase, another stress-activated protein kinase that can also stimulate c-jun gene expression, was not activated after NGF withdrawal. Consistent with Jun kinase activation, we found using a phospho-c-Jun-specific antibody that c-Jun was phosphorylated on serine 63 after NGF withdrawal. Furthermore, expression of a constitutively active form of MEK kinase 1 (MEKK1), which strongly activates the Jun kinase pathway, increased c-Jun protein levels and c-Jun phosphorylation and induced apoptosis in the presence of NGF. This death could be prevented by co-expression of SEKAL, a dominant negative mutant of SAPK/ERK kinase 1 (SEK1), an activator of Jun kinase that is a target of MEKK1. In contrast, expression of SEKAL alone did not prevent c-Jun expression, increases in c-Jun phosphorylation, or cell death after NGF withdrawal. Thus, activation of Jun kinase and increases in c-Jun phosphorylation and c-Jun protein levels occur at the same time after NGF withdrawal, but c-Jun levels and phosphorylation are regulated by an SEK1-independent pathway.


Subject(s)
Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Neurons/metabolism , Proto-Oncogene Proteins c-jun/biosynthesis , Superior Cervical Ganglion/metabolism , Animals , Animals, Newborn , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Nerve Growth Factors/deficiency , Neurons/enzymology , PC12 Cells , Phosphorylation , Protein Kinases/biosynthesis , Protein Kinases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/enzymology , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases
9.
J Neurosci ; 18(2): 751-62, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9425017

ABSTRACT

Cerebellar granule neurons die by apoptosis when deprived of survival signals. This death can be blocked by inhibitors of transcription or protein synthesis, suggesting that new gene expression is required. Here we show that c-jun mRNA and protein levels increase rapidly after survival signal withdrawal and that transfection of the neurons with an expression vector for a c-Jun dominant negative mutant protects them against apoptosis. Phosphorylation of serines 63 and 73 in the c-Jun transactivation domain is known to increase c-Jun activity. By using an antibody specific for c-Jun phosphorylated on serine 63, we show that this site is phosphorylated soon after survival signal withdrawal. To determine whether c-Jun phosphorylation is necessary for apoptosis, we have expressed c-Jun phosphorylation site mutants in granule neurons. c-Junasp, a constitutively active c-Jun mutant in which the known and potential serine and threonine phosphoacceptor sites in the transactivation domain have been mutated to aspartic acid, induces apoptosis under all conditions tested. In contrast, c-Junala, which cannot be phosphorylated because the same sites have been mutated to alanine, blocks apoptosis caused by survival signal withdrawal. Finally, we show that cerebellar granule neurons contain high levels of Jun kinase activity and low levels of p38 kinase activity, neither of which increases after survival signal withdrawal. Mitogen-activated protein kinase activity decreases under the same conditions. These results suggest that c-Jun levels and c-Jun phosphorylation may be regulated by novel mechanisms in cerebellar granule neurons.


Subject(s)
Apoptosis , Cerebellum/cytology , Mitogen-Activated Protein Kinases , Neurons/cytology , Proto-Oncogene Proteins c-jun/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA/metabolism , Dimerization , JNK Mitogen-Activated Protein Kinases , Neurons/metabolism , Phosphorylation , Potassium Chloride/metabolism , Rats , Rats, Sprague-Dawley , Serine/metabolism , Transcription, Genetic , Transcriptional Activation
11.
J Cell Biol ; 139(3): 809-15, 1997 Nov 03.
Article in English | MEDLINE | ID: mdl-9348296

ABSTRACT

The signaling pathways that mediate the ability of NGF to support survival of dependent neurons are not yet completely clear. However previous work has shown that the c-Jun pathway is activated after NGF withdrawal, and blocking this pathway blocks neuronal cell death. In this paper we show that over-expression in sympathetic neurons of phosphatidylinositol (PI) 3-kinase or its downstream effector Akt kinase blocks cell death after NGF withdrawal, in spite of the fact that the c-Jun pathway is activated. Yet, neither the PI 3-kinase inhibitor LY294002 nor a dominant negative PI 3-kinase cause sympathetic neurons to die if they are maintained in NGF. Thus, although NGF may regulate multiple pathways involved in neuronal survival, stimulation of the PI 3-kinase pathway is sufficient to allow cells to survive in the absence of this factor.


Subject(s)
Neurons/enzymology , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Superior Cervical Ganglion/enzymology , Superior Cervical Ganglion/physiology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Enzyme Activation , Nerve Growth Factors/deficiency , Nerve Growth Factors/physiology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/metabolism , Rats , Superior Cervical Ganglion/drug effects
12.
J Cell Sci ; 110 ( Pt 18): 2165-73, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9378766

ABSTRACT

In order to study the involvement of caspases in neuronal cell death, we have examined the effects of the viral caspase inhibitor p35 and peptide caspase inhibitors on sympathetic neurons isolated from the superior cervical ganglion (SCG). In these neurons, apoptosis can be induced by the withdrawal of nerve growth factor (NGF) and also by the addition of the kinase inhibitor staurosporine. p35 has been shown to be a broad spectrum inhibitor of the caspase family and promotes the survival of SCG neurons withdrawn from NGF. We show that p35 is also protective when apoptosis is induced by staurosporine. In addition, p35 inhibits a number of the morphological features associated with apoptosis, such as nuclear condensation, TUNEL labelling, and externalisation of phosphatidylserine. The tri-peptide caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp (O-methyl)-fluoromethylketone (zVAD-fmk) was effective at inhibiting NGF withdrawal-induced and staurosporine-induced apoptosis of SCG neurons. Two other peptide inhibitors, acetyl-Tyr-Val-Ala-Asp-aldehyde (Ac-YVAD-CHO) and acetyl-Asp-Glu-Ala-Asp-aldehyde (Ac-DEVD-CHO), also inhibited apoptosis induced by both means when microinjected into SCG neurons but peptides derived from the caspase cleavage site in p35 were not protective. We present data to suggest that apoptosis induced by separate death stimuli can result either in the activation of distinct caspases or in differences in the time of activation of the family members.


Subject(s)
Apoptosis/physiology , Caspases , Cysteine Endopeptidases/metabolism , Neurons/cytology , Neurons/enzymology , Superior Cervical Ganglion/cytology , Animals , Annexin A5/analysis , Apoptosis/drug effects , Biotin , Caenorhabditis elegans Proteins , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/physiology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Nerve Growth Factors/pharmacology , Neurons/chemistry , Peptide Fragments/analysis , Rats , Staining and Labeling , Staurosporine/pharmacology , Uracil Nucleotides
13.
J Cell Sci ; 110 ( Pt 14): 1603-13, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9247194

ABSTRACT

Endothelial cells provide a crucial interface between blood and tissue environments. Free diffusion of substances across endothelia is prevented by the endothelial tight junction, the permeability of which varies enormously depending on tissue. Endothelial cells of the blood-brain barrier possess tight junctions of severely limited permeability, whereas those of non-neural tissue are considerably leakier, but the molecular basis for this difference is not clear. Occludin is a major transmembrane protein localizing at the tight junction. In this study, we show, by immunocytochemistry, that occludin is present at high levels and is distributed continuously at cell-cell contacts in brain endothelial cells. In contrast, endothelial cells of non-neural tissue have a much lower expression of occludin, which is distributed in a discontinuous fashion at cell-cell contacts. The apparent differences in occludin expression levels were directly confirmed by immunoblotting. The differences in occludin protein were reflected at the message level, suggesting transcriptional regulation of expression. We also show that occludin expression is developmentally regulated, being low in rat brain endothelial cells at postnatal day 8 but clearly detectable at post-natal day 70. Our data indicate that regulation of occludin expression may be a crucial determinant of the tight junction permeability properties of endothelial cells in different tissues.


Subject(s)
Brain/blood supply , Membrane Proteins/analysis , Tight Junctions/chemistry , Tight Junctions/metabolism , Trans-Activators , Amino Acid Sequence , Animals , Astrocytes , Brain/cytology , Brain/growth & development , Cadherins/analysis , Capillaries/chemistry , Capillaries/metabolism , Cell Membrane Permeability/physiology , Cells, Cultured , Culture Media, Conditioned , Cytoskeletal Proteins/analysis , Endothelium/cytology , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Freeze Fracturing , Gene Expression Regulation, Developmental/physiology , Guinea Pigs , Membrane Proteins/genetics , Mice , Microscopy, Electron , Molecular Sequence Data , Occludin , Phosphoproteins/analysis , RNA, Messenger/analysis , Rats , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein , beta Catenin
14.
J Neurosci Res ; 48(6): 563-70, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9210526

ABSTRACT

Neuronal apoptosis is the subject of intense investigation and is beginning to be understood in some molecular detail. In the present study, we show that PC12 cells, like certain other cell types, redistribute phosphatidylserine (PS) from the inner leaflet to the outer leaflet of the plasma membrane early in the process of apoptosis. The externalised PS can be readily visualised by incubating intact cells with a fluorescent derivative of the protein annexin V. When apoptosis is blocked with an inhibitor of interleukin-1beta-converting-enzyme-like proteases, the increased annexin binding is also blocked. Fluorescent annexin V binding provides a rapid and convenient way to identify apoptotic neurones.


Subject(s)
Apoptosis , Cell Membrane/metabolism , PC12 Cells/metabolism , Phosphatidylserines/metabolism , Animals , Annexin A5/metabolism , Biomarkers , Caspase 1 , Cell Differentiation/drug effects , Cysteine Endopeptidases/physiology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , PC12 Cells/cytology , PC12 Cells/drug effects , Protease Inhibitors/pharmacology , Protein Binding , Rats , Staurosporine/pharmacology
15.
J Neurochem ; 68(4): 1598-605, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9084431

ABSTRACT

Proteases of the interleukin-1 beta converting enzyme (ICE) family have been implicated as mediators of apoptosis in several cell types. Here we report the ability of peptide inhibitors of ICE-like proteases to inhibit apoptosis of cultured cerebellar granule neurones caused by reduction of extracellular K+ levels and by the broad-spectrum protein kinase inhibitor staurosporine. Unlike apoptosis induced by K+ deprivation, staurosporine-induced neuronal death does not require new protein synthesis. The ICE-like protease inhibitor benzyloxycarbonyl-Val-Ala-Asp (O-methyl)fluoromethyl ketone (zVAD-fmk) was found to be extremely effective at preventing staurosporine-induced death of cerebellar granule neurones and yet was completely ineffective in preventing K+ deprivation-induced death. Staurosporine induced cleavage of the 116-kDa poly (ADP-ribose) polymerase enzyme, a substrate of ICE-like proteases, to the 85-kDa product, and this cleavage was also blocked by zVAD. By comparison, K+ deprivation led to the disappearance of the 116-kDa protein, with no detectable increase in level of the 85-kDa cleavage product. Taken together, these results imply the existence of divergent ICE-like protease pathways in a CNS model of neuronal apoptosis.


Subject(s)
Apoptosis/physiology , Cysteine Endopeptidases/metabolism , Neurons/cytology , Neurons/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase 1 , Cells, Cultured/cytology , Cerebellum/cytology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Interleukin-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology
16.
Development ; 124(6): 1239-49, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9102310

ABSTRACT

The Bcl-2 and Bcl-x proteins suppress programmed cell death, whereas Bax promotes apoptosis. We investigated the pattern of expression of Bcl-2, Bax and Bcl-x during neuronal differentiation and development. All three proteins were widely expressed in neonatal rats but, in the adult, Bax levels were 20- to 140-fold lower in the cerebral cortex, cerebellum and heart muscle, whereas Bcl-x was not downregulated in any of the tissues examined. In the cerebral cortex and cerebellum, the decrease in Bax levels occurred after the period of developmental cell death. Further, microinjection of a Bax expression vector into cultured sympathetic neurons, which depend on nerve growth factor for survival, induced apoptosis in the presence of survival factor and increased the rate of cell death after nerve growth factor withdrawal. This effect could be blocked by co-injection of an expression vector for Bcl-xL or for the baculovirus p35 protein, an inhibitor of caspases (ICE-like proteases). These results suggest that, during development, the sensitivity of neurons to signals that induce apoptosis may be regulated by modulating Bax levels and that Bax-induced death requires caspase activity.


Subject(s)
Aging/physiology , Apoptosis , Brain/growth & development , Gene Expression Regulation, Developmental , Neurons/physiology , Proto-Oncogene Proteins/biosynthesis , Amino Acid Sequence , Animals , Animals, Newborn , Brain/metabolism , Cell Differentiation , Cell Survival , Cerebellum/growth & development , Cerebral Cortex/growth & development , Humans , Molecular Sequence Data , Nerve Growth Factors , Neurons/cytology , Organ Specificity , PC12 Cells , Peptide Fragments/chemistry , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein , bcl-X Protein
17.
J Neurochem ; 68(3): 991-1000, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048744

ABSTRACT

Brain capillary endothelial cells are coupled by a continuous belt of complex high-electrical-resistance tight junctions that are largely responsible for the blood-brain barrier. We have investigated mechanisms regulating tight junction permeability in brain endothelial cells cultured to maintain high-resistance junctions. The phospholipid lysophosphatidic acid (LPA) was found to cause a rapid, reversible, and dose-dependent decrease in transcellular electrical resistance in brain endothelial cells. LPA also increased the paracellular flux of sucrose, which, together with the resistance decrease, indicated increased tight junction permeability. Activation of protein kinase C attenuated the effect of LPA, suggesting that it was mediated by activation of a signalling pathway. LPA did not cause any obvious relocalization of adherens junction- or tight junction-associated proteins. However, it did stimulate the formation of stress fibres, the recruitment of focal adhesion components, and the appearance of tyrosine phosphorylated protein at focal contacts. Our study shows that LPA is a modulator of tight junction permeability in brain endothelial cells in culture and raises the possibility that it triggers blood-brain barrier permeability changes under (patho)physiological conditions.


Subject(s)
Capillary Permeability/drug effects , Cerebrovascular Circulation/drug effects , Endothelium, Vascular/metabolism , Lysophospholipids/pharmacology , Tight Junctions/metabolism , Animals , Cells, Cultured , Electric Impedance , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Immunohistochemistry , Mice , Phosphorylation , Signal Transduction , Swine , Tyrosine/metabolism
18.
Br Med Bull ; 53(3): 617-31, 1997.
Article in English | MEDLINE | ID: mdl-9374041

ABSTRACT

Apoptosis is recognised increasingly as a prominent event in nervous system development and disease. This form of death appears to obey the same rules in neurones as in other cells, in that it is initiated by similar extracellular perturbations and distinguished by similar morphological and biochemical changes. When neurones die after survival factor withdrawal, gene transcription is important, with the transcription factor c-jun and the cytoplasmic signalling cascade that regulates it being particularly significant in at least some types of cells. However, death can be induced in a transcription-independent manner by agents such as staurosporine. Both types of death involve activation of members of the ICE family of proteases but, surprisingly, the particular protease involved seems to depend very much on the manner in which death is initiated.


Subject(s)
Apoptosis/physiology , Neurons/cytology , Apoptosis/genetics , Cell Culture Techniques , Cerebrovascular Disorders/pathology , Cytoplasm/metabolism , Humans , Necrosis , Neurodegenerative Diseases/pathology , Proto-Oncogene Proteins c-bcl-2/physiology
19.
J Biol Chem ; 272(50): 31894-901, 1997 Dec 12.
Article in English | MEDLINE | ID: mdl-9395537

ABSTRACT

Protein kinase C signaling pathways have been implicated in the disruption of intercellular junctions, but mechanisms are not clear. p100 and p120 are members of the Armadillo family of proteins and are localized to cellular adherens junctions. In strain I Madin-Darby canine kidney cells, protein kinase C activation leads to disruption of tight junctions and an increase in permeability of cell monolayers. We show that this permeability increase is accompanied by dephosphorylation of p100/p120 on serine and threonine residues. The dephosphorylation of these proteins can also be induced by the kinase inhibitors staurosporine, KT5926, and Gö 6976. Treatment of cells with phosphatase inhibitors induced hyperphosphorylation of p100 and p120. Thus, p100 and p120 participate in a regulatable cycle of serine/threonine phosphorylation and dephosphorylation. Protein kinase C must act, directly or indirectly, by perturbing this phosphorylation cycle, by inhibition of a p100/p120 kinase and/or activation of a phosphatase. These data clearly show that p100 and p120 are targets of a novel protein kinase C signaling pathway. Dephosphorylation of these proteins precedes the permeability increase across epithelial cell monolayers seen in response to phorbol esters, raising the possibility that this pathway may play a role in the modulation of intercellular junctions.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/enzymology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Animals , Catenins , Dogs , Enzyme Activation , Models, Chemical , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Serine/metabolism , Threonine/metabolism , Delta Catenin
20.
Curr Opin Neurobiol ; 6(5): 622-7, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8937826

ABSTRACT

The blood-brain barrier regulates the movement of molecules and cells between the circulation and the CNS. Modulation of this barrier may be critical in the aetiology of various CNS pathologies. Endothelial cell tight junctions are an essential part of the barrier, and recent advances have been made in understanding how specific intracellular signalling events regulate cell-cell adhesion and tight-junction permeability.


Subject(s)
Blood-Brain Barrier , Brain/physiology , Cell Adhesion , Endothelium, Vascular/physiology , Animals , Brain/pathology , Brain/physiopathology , Cytoskeleton/physiology , Models, Neurological , Signal Transduction , Tight Junctions/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...