Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 4(9): 1989-96, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16961606

ABSTRACT

BACKGROUND: A significant need exists for new chronic oral anticoagulation therapies to replace warfarin. Previous studies have shown that beta-D-xylosides, which prime glycosaminoglycan (GAG) synthesis, have antithrombin and antithrombotic activity. In the following report, a new orally active beta-D-xyloside (odiparcil) has been characterized in a rat model of venous thrombosis and its efficacy and bleeding liability compared to warfarin. Additionally, studies were conducted to investigate odiparcil's ex vivo antithrombin and antiplatelet activity, and also to explore the potential utility of protamine sulfate as a neutralizing agent. METHODS AND RESULTS: In vivo thrombosis studies were conducted in a rat inferior vena cava model, and bleeding studies in a rat tail transection model. Following oral dosing, warfarin and odiparcil produced dose-related suppression of thrombus formation. A therapeutically relevant dose of warfarin in this model (international normalized ratio; INR 3.0) achieved approximately 65% inhibition of thrombus formation. Warfarin caused dose-related significant increases in bleeding indices. Odiparcil antithrombotic activity was limited by its mechanism to a maximum suppression of thrombus formation of 65-70%, and did not prolong bleeding indices. Additionally, odiparcil-induced heparin cofactor II (HCII)-dependent antithrombin activity was shown to be a function of dermatan sulfate-like GAG production. Other than thrombin-related effects, no odiparcil effects on platelet function were observed. In antidote studies, it was demonstrated that odiparcil-induced antithrombotic activity could be partially neutralized by protamine sulfate. CONCLUSIONS: These experiments suggest that an antithrombotic approach based upon xyloside induction of circulating GAGs may have the potential to approximate the efficacy of warfarin and yet with a reduced risk to hemostasis.


Subject(s)
Glycosides/therapeutic use , Venous Thrombosis/drug therapy , Warfarin/therapeutic use , Animals , Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glycosaminoglycans/blood , Glycosides/adverse effects , Hemorrhage/chemically induced , Heparin Cofactor II , Protamines/therapeutic use , Rats , Vena Cava, Inferior , Warfarin/adverse effects
2.
J Thromb Haemost ; 4(4): 882-90, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16634759

ABSTRACT

P2X1 receptors are ATP-gated channel demonstrated to be involved in multiple platelet responses, although in vitro analysis has been complicated by the effects of rapid desensitization. To further investigate potential roles of P2X1 receptors in platelet activation, the current study employed methods which maximally preserved P2X1 functionality. In preliminary in vivo studies, P2X1-deficiency reduced thrombus formation following the laser-induced, but not FeCl3-induced injury. Given the multiple potential mechanisms involved in thrombus formation in vivo, including tissue-factor/thrombin generation pathways, subsequent studies were designed to investigate the effects of P2X1 inhibition or stimulation on platelet activation in vitro; specifically, the interaction of P2X1 with thrombin receptor stimulation. Aggregation initiated by low/threshold levels of a protease-activated receptor (PAR)4 agonist was reduced in P2X1-deficient murine platelets, and inhibition of P2X1 in wild-type platelets similarly reduced PAR4-mediated aggregation. In human platelets, aggregation to low/threshold stimulation of PAR1 was inhibited with the P2X1 antagonist MRS2159. In addition, P2X1 stimulation primed human platelet responses, such that subsequent sub-threshold PAR1 responses were converted into significant aggregation. Selective ADP receptor inhibitors attenuated P2X1-mediated priming, suggesting that the synergy between P2X1 and sub-threshold PAR1 stimulation was in part because of enhanced granular release of ADP. Overall, the present study defines a novel interaction between platelet P2X1 and thrombin receptors, with P2X1 functioning to amplify aggregation responses at low levels of thrombin receptor stimulation.


Subject(s)
Platelet Aggregation , Receptors, Purinergic P2/metabolism , Receptors, Thrombin/metabolism , Animals , Blood Platelets/metabolism , Chlorides , Ferric Compounds/pharmacology , Humans , Lasers , Mice , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X , Species Specificity , Thrombosis/metabolism
3.
J Thromb Haemost ; 1(12): 2626-35, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14675100

ABSTRACT

The platelet P2X1 purinergic receptor is a ligand-gated ion channel that responds to ATP. The precise role of P2X1 in platelet function is unknown, though stimulation with the P2X1 agonist alpha,beta-Me-ATP is known to result in platelet shape change through elevation of calcium levels. The aim of the present study was to examine further the effects of P2X1 stimulation on platelet activation. Stimulation of P2X1 with alpha,beta-Me-ATP resulted in shape change and small aggregate formation in heparin-anticoagulated platelet preparations. Given the ability of heparin to potentiate platelet activation, subsequent experiments were performed in hirudin. In these platelet preparations, aggregate formation in response to alpha,beta-Me-ATP alone was less than that observed in heparin; however, alpha,beta-Me-ATP significantly potentiated platelet aggregate formation when added in conjunction with other weak platelet agonists [epinephrine or thrombopoietin (TPO)]. Platelet aggregate formation was confirmed by single platelet loss (microaggregate formation), microscopy, and light transmittance studies. Further, the P2X1 antagonist MRS-2159 inhibited platelet shape change and aggregation responses induced by alpha,beta-Me-ATP. Overall, this study demonstrates that P2X1 stimulation can induce/potentiate platelet activation in combination with other platelet agonists. These results are the first demonstration of platelet aggregation mediated through direct P2X1 stimulation, supporting a role for this receptor in regulating platelet activation.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Platelet Activation , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/pharmacology , Calcium Signaling , Cell Size/drug effects , Epinephrine/pharmacology , Heparin/pharmacology , Humans , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Function Tests , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2X , Thrombopoietin/pharmacology
5.
J Neurosci ; 21(10): 3303-11, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331359

ABSTRACT

Oxidant-induced neuronal apoptosis has been shown to involve potassium and zinc dysregulation, energetic dysfunction, activation of stress-related kinases, and caspase cleavage. The temporal ordering and interdependence of these events was investigated in primary neuronal cultures exposed to the sulfhydryl oxidizing agent 2,2'-dithiodipyridine (DTDP), a compound that induces the intracellular release of zinc. We previously observed that tetraethylammonium (TEA), high extracellular potassium, or cysteine protease inhibitors block apoptosis induced by DTDP. We now report that both p38 and extracellular signal-regulated kinase phosphorylation are evident in neuronal cultures within 2 hr of a brief exposure to 100 microm DTDP. However, only p38 inhibition is capable of blocking oxidant-induced toxicity. Cyclohexamide or actinomycin D does not attenuate DTDP-induced cell death, suggesting that posttranslational modification of existing targets, rather than transcriptional activation, is responsible for the deleterious effects of p38. Indeed, an early robust increase in TEA-sensitive potassium channel currents induced by DTDP is attenuated by p38 inhibition but not by caspase inhibition. Moreover, we found that activation of p38 is required for caspase 3 and 9 cleavage, suggesting that potassium currents enhancement is required for caspase activation. Finally, we observed that DTDP toxicity could be blocked with niacinamide or benzamide, inhibitors of poly (ADP-ribose) synthetase. Based on these findings, we conclude that oxidation of sulfhydryl groups on intracellular targets results in intracellular zinc release, p38 phosphorylation, enhancement of potassium currents, caspase cleavage, energetic dysfunction, and translationally independent apoptotic cell death.


Subject(s)
Caspases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Oxidants/pharmacology , Potassium Channels/metabolism , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Benzamides/pharmacology , Caspase Inhibitors , Cells, Cultured , Disulfides/antagonists & inhibitors , Disulfides/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/cytology , Neurons/drug effects , Niacinamide/pharmacology , Oxidants/antagonists & inhibitors , Oxidative Stress/drug effects , Patch-Clamp Techniques , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerase Inhibitors , Protein Synthesis Inhibitors/pharmacology , Rats , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/antagonists & inhibitors , Sulfhydryl Reagents/pharmacology , Zinc/metabolism , p38 Mitogen-Activated Protein Kinases
6.
Brain Res ; 892(1): 70-7, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11172750

ABSTRACT

The aim of the present study was to evaluate p38 MAPK activation following focal stroke and determine whether SB 239063, a novel second generation p38 inhibitor, would directly attenuate early neuronal injury. Following permanent middle cerebral artery occlusion (MCAO), brains were dissected into ischemic and non-ischemic cortices and Western blots were employed to measure p38 MAPK activation. Neurologic deficit and MR imaging were utilized at various time points following MCAO to monitor the development and resolution of brain injury. Following MCAO, there was an early (15 min) activation of p38 MAPK (2.3-fold) which remained elevated up to 1 h (1.8-fold) post injury compared to non-ischemic and sham operated tissue. Oral SB 239063 (5, 15, 30, 60 mg/kg) administered to each animal 1 h pre- and 6 h post MCAO provided significant (P<0.05) dose-related neuroprotection reducing infarct size by 42, 48, 29 and 14%, respectively. The most effective dose (15 mg/kg) was further evaluated in detail and SB 239063 significantly (P<0.05) reduced neurologic deficit and infarct size by at least 30% from 24 h through at least 1 week. Early (i.e. observed within 2 h) reductions in diffusion weighted imaging (DWI) intensity following treatment with SB 239063 correlated (r=0.74, P<0.01) to neuroprotection seen up to 7 days post stroke. Since increased protein levels for various pro-inflammatory cytokines cannot be detected prior to 2 h in this stroke model, the early improvements due to p38 inhibition, observed using DWI, demonstrate that p38 inhibition can be neuroprotective through direct effects on ischemic brain cells, in addition to effects on inflammation.


Subject(s)
Cerebral Infarction/prevention & control , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Ischemic Attack, Transient/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Pyrimidines/pharmacology , Animals , Cell Death/drug effects , Cerebral Infarction/pathology , Ischemic Attack, Transient/pathology , Male , Middle Cerebral Artery , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/pathology , Rats , Rats, Inbred SHR , p38 Mitogen-Activated Protein Kinases
7.
Med Res Rev ; 21(2): 129-45, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11223862

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are involved in many cellular processes. The stress-activated MAPK, p38, has been linked to inflammatory cytokine production and cell death following cellular stress. Here, we demonstrate focal ischemic stroke-induced p38 enzyme activation (i.e., phosphorylation) in the brain. The second generation p38 MAPK inhibitor SB 239063 was identified to exhibit increased kinase selectivity and improved cellular and in vivo activity profiles, and thus was selected for evaluation in two rat models of permanent focal ischemic stroke. SB 239063 was administered orally pre- and post-stroke and intravenously post-stroke. Plasma concentration levels were achieved in excess of those that effectively inhibit p38 activity. In both moderate and severe stroke, SB 239063 reduced infarct size by 28-41%, and neurological deficits by 25-35%. In addition, neuroprotective plasma concentrations of SB 239063 that reduced p38 activity following stroke also reduced the stroke-induced expression of IL-1beta and TNFalpha (i.e., cytokines known to contribute to stroke-induced brain injury). SB 239063 also provided direct protection of cultured brain tissue to in vitro ischemia. This robust SB 239063-induced neuroprotection emphasizes a significant opportunity for targeting MAPK pathways in ischemic stroke injury, and also suggests that p38 inhibition be evaluated for protective effects in other experimental models of nervous system injury and neurodegeneration.


Subject(s)
Brain Ischemia/drug therapy , Imidazoles/therapeutic use , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Pyrimidines/therapeutic use , Animals , Brain Ischemia/metabolism , Cells, Cultured , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacokinetics , Interleukin-1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Rats , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
8.
Drug Discov Today ; 6(2): 85-91, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11166256

ABSTRACT

Caspases are essential for apoptosis. A crucial question regarding the role(s) of these proteases is whether the selective inhibition of an effector caspase will prevent cell death. We have identified potent, selective non-peptide inhibitors of the effector caspases 3 and 7. Apoptosis can be inhibited and cell functionality maintained using an inhibitor selective for caspases 3 and 7. This has important therapeutic implications and the potential to generate novel anti-apoptotic strategies in diseases that involve dysregulated apoptosis.

9.
J Pharmacol Exp Ther ; 296(2): 312-21, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160612

ABSTRACT

The stress-activated mitogen-activated protein kinase (MAPK) p38 has been linked to the production of inflammatory cytokines/mediators/inflammation and death/apoptosis following cell stress. In these studies, a second-generation p38 MAPK inhibitor, SB 239063 (IC(50) = 44 nM), was found to exhibit improved kinase selectivity and increased cellular (3-fold) and in vivo (3- to 10-fold) activity over first-generation inhibitors. Oral SB 239063 inhibited lipopolysaccharide-induced plasma tumor necrosis factor production (IC(50) = 2.6 mg/kg) and reduced adjuvant-induced arthritis (51% at 10 mg/kg) in rats. SB 239063 reduced infarct volume (48%) and neurological deficits (42%) when administered orally (15 mg/kg, b.i.d.) before moderate stroke. Intravenous SB 239063 exhibited a clearance of 34 ml/min/kg, a volume of distribution of 3 l/kg, and a plasma half-life of 75 min. An i.v. dosing regimen that provided effective plasma concentrations of 0.38, 0.75, or 1.5 microg/ml (i.e., begun 15 min poststroke and continuing over the initial 6-h p38 activation period) was used. Significant and dose-proportional brain penetration of SB 239063 was demonstrated during these infusion periods. In both moderate and severe stroke, intravenous SB 239063 produced a maximum reduction of infarct size by 41 and 27% and neurological deficits by 35 and 33%, respectively. No effects of the drug were observed on cerebral perfusion, hemodynamics, or body temperature. Direct neuroprotective effects from oxygen and glucose deprivation were also demonstrated in organotypic cultures of rat brain tissue. This robust in vitro and in vivo SB 239063-induced neuroprotection emphasizes the potential role of MAPK pathways in ischemic stroke and also suggests that p38 inhibition warrants further study, including protection in other models of nervous system injury and neurodegeneration.


Subject(s)
Brain/pathology , Enzyme Inhibitors/therapeutic use , Imidazoles/therapeutic use , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Pyrimidines/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Body Temperature/drug effects , Cerebrovascular Circulation/drug effects , Hemodynamics/drug effects , Hippocampus/pathology , Inflammation/pathology , Inflammation/prevention & control , Organ Culture Techniques , Pyridines/therapeutic use , Rats , Rats, Inbred Lew , Rats, Inbred SHR , p38 Mitogen-Activated Protein Kinases
10.
Expert Opin Emerg Drugs ; 6(1): 81-94, 2001 Apr.
Article in English | MEDLINE | ID: mdl-15989498

ABSTRACT

Apoptotic neuronal cell death has been demonstrated to occur in the central nervous system (CNS), following both acute injury and during chronic neurodegenerative conditions. Currently, the majority of experimental evidence for a role of caspases in CNS damage has been established following acute neuronal insults, including ischaemic stroke, traumatic brain injury and spinal cord injury. In vitro and in vivo models have been used to demonstrate caspase activation, and treatment with available caspase inhibitors can provide significant protection. Overall, acute neuronal injury represents a major unmet medical need and caspase inhibitors may be an attractive approach to preserve neuronal function by extending the therapeutic window and providing long-term neuroprotection. Currently, several inhibitors are in preclinical drug development and this review summarises recent advances in the development of novel caspase inhibitors for the treatment of acute neuronal injury.

11.
Neuroreport ; 11(17): 3719-23, 2000 Nov 27.
Article in English | MEDLINE | ID: mdl-11117479

ABSTRACT

PEP-19 is a calmodulin-regulatory protein found specifically within neurons, though cellular functions of this protein have not been determined. In an effort to define potential effects of PEP-19, PC12 cell lines expressing this protein were generated and subjected to apoptotic stimuli. As measured by LDH release, cell death in PEP-19 expressing cells was 2- to 5-fold less following u.v. irradiation, and 2- to 4-fold less following staurosporine treatment than controls. Additionally, PEP-19-expressing cells displayed decreased DNA ladder formation, chromatin and condensation, caspase activation following staurosporine treatment. Overall, these results demonstrate that PEP-19 can inhibit apoptotic processes in PC12 cells, suggesting a potential regulatory mechanism for pathways leading to cell death.


Subject(s)
Apoptosis/drug effects , Nerve Tissue Proteins/physiology , Neurons/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin-Binding Proteins , Enzyme Activation , L-Lactate Dehydrogenase/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/radiation effects , PC12 Cells , Rats , Staurosporine/pharmacology , Ultraviolet Rays
12.
J Cereb Blood Flow Metab ; 20(8): 1197-204, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10950380

ABSTRACT

Increasing evidence supports a role for oxidative stress, proinflammatory cytokines, and apoptosis in the pathophysiology of focal ischemic stroke. Previous studies have found that the multi-action drug, carvedilol, is a mixed adrenergic antagonist, and that it behaves as an antioxidant and inhibits apoptosis. In the current study, the authors investigated whether carvedilol provides protection in focal cerebral ischemia and whether this protection is associated with reduced apoptosis and the downregulation of the inflammatory cytokines, tumor necrosis factor-alpha (TNF-alpha) and interleukin- 1beta (IL-1beta). Male Sprague-Dawley rats were subjected to transient middle cerebral artery occlusion (MCAO) by an intraluminal filament technique. Carvedilol (1, 3, and 10 mg/kg) was injected daily subcutaneously 2 or 4 days before the induction of ischemia. Neurologic scores, infarct volumes, TUNEL staining, and mRNA levels of TNF-alpha and IL-1beta were assessed at 24 hours reperfusion. The effect of carvedilol on microvascular cortical perfusion was studied with continuous laser-Doppler flowmetry. Twenty-four hours after MCAO, carvedilol at all three doses reduced infarct volumes by at least 40% and reduced neurologic deficits on average by 40% compared with vehicle-treated controls when given 2 or 4 days before the induction of ischemia. This protection was not mediated by changes in temperature or blood flow. Treatment with all three dose regimens resulted in fewer TUNEL positive cells compared with controls. At 24 hours reperfusion, carvedilol decreased TNF-alpha and IL-1beta expression by 40% to 50% in the ipsilateral ischemic cortex compared with the contralateral controls. The results of the current study indicate that carvedilol is neuroprotective in focal cerebral ischemia and may protect the ischemic brain by inhibiting apoptosis and attenuating the expression of TNF-alpha and IL-1beta.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Brain/drug effects , Brain/pathology , Carbazoles/pharmacology , Ischemic Attack, Transient/pathology , Neuroprotective Agents/pharmacology , Propanolamines/pharmacology , Stroke/pathology , Animals , Apoptosis , Carvedilol , Cerebral Cortex/blood supply , Cerebral Infarction/pathology , Cerebrovascular Circulation , Interleukin-1/metabolism , Male , Nervous System/physiopathology , Rats , Rats, Sprague-Dawley , Stroke/physiopathology , Tumor Necrosis Factor-alpha/metabolism
13.
Neurosci Lett ; 282(3): 189-92, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10717423

ABSTRACT

The aim of the present study was to quantitate the temporal changes in protein concentration for interleukin (IL)-1alpha, IL-1beta, IL-1ra, and IL-6 from 1 h to 15 days following focal ischemia. Protein expression was evaluated by enzyme-linked immunosorbent assay utilizing newly available rat antibodies. There were no detectable basal levels of IL-1alpha, 1L-1beta, or IL-6 in the sham-operated or non-ischemic control cortex. IL-1beta (increased significantly (P<0.05) as early as 4 h and peaked at 3 to 5 days. IL-1alpha (increased significantly (P<0.05) at 3 days. IL-6 increased early and peaked at 24 h (P<0.05). IL-1ra increased significantly (P<0.05) over basal levels from 12 h to 5 days. The present study provides the first quantitative determination of interleukin protein concentrations in the rat brain following focal stroke and demonstrates that this technology is now available for mechanistic studies in neuroprotection.


Subject(s)
Interleukins/metabolism , Ischemic Attack, Transient/metabolism , Animals , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/metabolism , Interleukin-6/metabolism , Male , Rats , Rats, Inbred SHR , Sialoglycoproteins/metabolism
14.
J Cereb Blood Flow Metab ; 20(1): 15-20, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10616788

ABSTRACT

A short duration of ischemia (i.e., ischemic preconditioning) results in significant brain protection to subsequent severe ischemic insult. Because previous studies suggest that tumor necrosis factor-alpha (TNF-alpha) plays a role in both promoting ischemic damage and neuroprotection, the present work aimed to evaluate the expression of TNF-alpha mRNA in an established model of ischemic preconditioning using a transient 10-minute occlusion of the middle cerebral artery. Because the level of TNF-alpha mRNA expression in the brain was too low to be consistently detected by Northern technique, a real-time polymerase chain reaction method was applied to quantitate the absolute copy number of TNF-alpha transcript in rat brain after the preconditioning procedure. TNF-alpha mRNA was induced in the ipsilateral cortex as early as 1 hour (27 +/- 1 copies of mRNA per microgram of tissue compared to 11 +/- 3 copies in sham-operated samples) after preconditioning, reached a peak level at 6 hours (49 +/- 10 copies of transcript, n = 4, P < 0.01), and persisted up to 2 days. These data not only demonstrate the utility of real-time polymerase chain reaction for sensitive and accurate measurement of mRNA expression in normal and injured tissues but also suggest a potential role of TNF-alpha in the phenomenon of ischemic preconditioning.


Subject(s)
Brain/blood supply , Brain/metabolism , Ischemic Preconditioning , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Cloning, Molecular , Computer Systems , DNA/genetics , Plasmids , Polymerase Chain Reaction/methods , Rats , Ribosomal Proteins/genetics , Time Factors
15.
Mol Neurobiol ; 22(1-3): 99-113, 2000.
Article in English | MEDLINE | ID: mdl-11414283

ABSTRACT

Neuromodulin (GAP-43), neurogranin (RC3), and PEP-19 are small acid-stable proteins that bind calcium-poor calmodulin through a loosely conserved IQ-motif. Even though these proteins have been known for many years, much about their function in cells is not understood. It has recently become appreciated that calmodulin activity in cells is tightly controlled and that pools of otherwise free calmodulin are sequestered so as to restrict its availability for activating calcium/calmodulin-dependent enzymes. Neuromodulin, neurogranin, and PEP-19 appear to be major participants in this type of regulation. One way in which they do this is by providing localized increases in the concentration of calmodulin in cells so that the maximal level of target activation is increased. Additionally, they can function as calmodulin antagonists by directly inhibiting the association of calcium/calmodulin with enzymes and other proteins. Although neuromodulin, neurogranin, and PEP-19 were early representatives of the small IQ-motif-containing protein family, newer examples have come to light that expand the number of cellular systems through which the IQ-peptide/calmodulin interaction could regulate biological processes including gene transcription. It is the purpose of this review to examine the behavior of neuromodulin, neurogranin, and PEP-19 in paradigms that include both in vitro and in situ systems in order to summarize possible biological consequences that are linked to the expression of this type of protein. The use of protein:protein interaction chromatography is also examined in the recovery of a new calmodulin-binding peptide, CAP-19 (ratMBF1). Consistent with earlier predictions, at least one function of small IQ-motif proteins appears to be that they lessen the extent to which calcium-calmodulin-dependent enzymes become or stay activated. It also appears that these polypeptides can function to selectively inhibit activation of intracellular targets by some agonists while simultaneously permitting activation of these same targets by other agonists. Much of the mechanism for how this occurs is unknown, and possible explanations are examined. One of the biological consequences for a cell that expresses a calmodulin-regulatory protein could be an increased resistance to calcium-mediated toxicity. This possibility is examined for cells expressing PEP-19 and both anatomical and cell-biological data is described. The study of IQ-motif-containing small proteins has stimulated considerable thought as to how calcium signaling is refined in neurons. Current evidence suggests that signaling through calmodulin is not a fulminating and homogenous process but a spatially limited and highly regulated one. Data from studies on neuromodulin, neurogranin, and PEP-19 suggest that they play an important role in establishing some of the processes by which this regulation is accomplished.


Subject(s)
Calcium Signaling/physiology , Calmodulin-Binding Proteins/physiology , Calmodulin/physiology , GAP-43 Protein/physiology , Nerve Tissue Proteins/physiology , Alzheimer Disease/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Brain Chemistry , Calcium/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/chemistry , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Enzyme Activation/drug effects , GAP-43 Protein/chemistry , GAP-43 Protein/pharmacology , Homeostasis , Humans , Huntington Disease/metabolism , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nerve Degeneration , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/pharmacology , Neurogranin , Neurons/drug effects , Neurons/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , PC12 Cells/drug effects , PC12 Cells/enzymology , Phosphorylation , Protein Processing, Post-Translational , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
16.
J Biol Chem ; 273(52): 35222-7, 1998 Dec 25.
Article in English | MEDLINE | ID: mdl-9857061

ABSTRACT

NFB42 (neural F Box 42 kDa) is a novel gene product that is highly enriched in the nervous system. Its predicted protein contains an F box, a motif recently shown to couple cell cycle regulation to the proteasome pathway (Bai, C., Sen, P., Hofmann, K., Ma, L., Goebl, M., Harper, J. W., and Elledge, S. (1996) Cell 86, 263-274). NFB42 mRNA and protein are expressed in all major areas of the adult rat brain but are not detected in non-neural tissues. NFB42 protein is localized primarily to the cytoplasm of neurons and does not appear to be present in glia. The presence of an F box in NFB42 suggests that it may be involved in cell cycle regulation; however, its expression in postmitotic neurons indicates that it is not involved in regulating typical cell cycle events. In an initial attempt to characterize the function of this protein, NFB42 was transfected into N1E-115 neuroblastoma and Chinese hamster ovary cells. The expression of full-length NFB42, but not an F box deletion mutant, inhibits proliferation in both cell lines. Additional experiments demonstrate that NFB42 interacts with Skp1p, a component of the proteasome pathway, and deletion of the F box also inhibits this interaction. Overall, the expression pattern of NFB42, along with the presence of an F box domain and the ability to inhibit growth, suggests that it may play a role in maintaining neurons in a postmitotic state.


Subject(s)
Cell Cycle Proteins/isolation & purification , Growth Inhibitors/isolation & purification , Nerve Tissue Proteins/isolation & purification , Neurons/chemistry , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/pharmacology , Cloning, Molecular , Cricetinae , DNA, Complementary/genetics , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/cytology , PC12 Cells , Protein Binding , Rats , Recombinant Proteins , S-Phase Kinase-Associated Proteins , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured
17.
J Biol Chem ; 273(36): 23517-23, 1998 Sep 04.
Article in English | MEDLINE | ID: mdl-9722590

ABSTRACT

Nerve growth factor treatment of PC12 cells results in neuronal differentiation, a process accompanied by induction of the Cdk inhibitor p21(WAF1). To determine the role of p21 in differentiation, PC12 clones containing an inducible p21 construct were utilized to induce growth arrest. Expression of p21 led to accumulation of cyclins D1 and E and to a decrease in cyclins A and B. Levels of Cdc2 and Cdk4 also decreased after p21 induction. Initially, thymidine incorporation into DNA was dramatically inhibited; however, low levels of incorporation were observed during prolonged p21 expression. Fluorescence-activated cell sorter analysis revealed that this low level of DNA synthesis resulted in the generation of polyploid cells. Results from Western blots were consistent with phosphorylation of p21 protein coincident with the resumption of DNA synthesis. Finally, treatment of p21-arrested populations with epidermal growth factor, a known PC12 mitogen, resulted in neurite extension, a key feature of neuronal differentiation. Overall, cell cycle changes following p21 overexpression in PC12 cells closely mimic distinctive events previously shown to occur during differentiation. These results suggest that the mechanism by which nerve growth factor induces the many cellular changes associated with growth arrest during differentiation is through p21(WAF1) induction.


Subject(s)
Cyclins/biosynthesis , Nerve Growth Factors/pharmacology , Neurons/cytology , Proto-Oncogene Proteins , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle , Cell Differentiation , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Epidermal Growth Factor/pharmacology , Neurites , PC12 Cells , Phosphorylation , Rats
18.
Oncogene ; 16(4): 443-51, 1998 Jan 29.
Article in English | MEDLINE | ID: mdl-9484833

ABSTRACT

p21WAF1 cyclin-dependent kinase inhibitor has been implicated in the control of proliferation, differentiation, and death in various cell lines. To further examine p21 regulation of the transitions between these cellular processes, an inducible p21 vector (lac operon system) was transfected into the rat pheochromocytoma (PC12) neural cell line. Induction of p21 led to permanent growth arrest, as evidenced by cell counts, FACS analysis, and thymidine incorporation. This arrest was maintained, even after removal of the inducing signal (IPTG). Northern analysis revealed that endogenous p21 mRNA increased following IPTG removal, which may be responsible for the continued growth arrest despite the decrease in ectopic p21 expression. p21 overexpression did not directly lead to a differentiated phenotype; however, differentiation in response to nerve growth factor (NGF) was greatly accelerated. To examine effects on cell death, and specifically test the hypothesis that apoptosis caused by withdrawal of trophic support results from inappropriate entry into cell cycle, serum was removed from proliferating and p21-arrested PC12 cells. The rate of apoptotic death was not affected by p21, nor was it effective in altering the extent of death following other apoptotic stimuli.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cyclins/physiology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/genetics , Isopropyl Thiogalactoside/pharmacology , Nerve Growth Factors/pharmacology , Neurons/cytology , PC12 Cells , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...