Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters











Publication year range
1.
CNS Neurol Disord Drug Targets ; 11(6): 705-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22963440

ABSTRACT

Purine nucleotides are well established as extracellular signaling molecules. P2X7 receptors (P2X7Rs) are members of the family of ionotropic ATP-gated receptors. Their activity can be found in a limited number of cell types, but is readily detectable in cells of hemopoietic lineage including macrophages, microglia, and certain lymphocytes, and mediates the influx of Ca2+ and Na+ as well as the release of pro-inflammatory cytokines. Amongst P2X receptors, P2X7Rs behave as a bifunctional molecule. The binding of ATP induces within milliseconds the opening of a channel selective for small cations, and within seconds a larger pore opens which allows permeation by molecules with a mass of up to 900 Da. In humans at least, the P2RX7 gene is highly polymorphic, and genetic differences within P2X7R affect receptor pore formation and channel function. ATP can act as a neurotransmitter, while the presence of P2X7Rs on immune cells suggests that they also regulate immune function and inflammatory responses. In addition, activation of the P2X7R has dramatic cytotoxic properties. The role of extracellular ATP and purinoceptors in cytokine regulation and neurological disorders is, in fact, the focus of a rapidly expanding area of research. P2X7Rs may affect neuronal cell death by regulating the processing and release of interleukin-1ß, a key mediator in neurodegeneration, chronic inflammation, and chronic pain. Activation of P2X7Rs provides an inflammatory stimulus, and P2X7R-deficient mice display a marked attenuation of inflammatory responses, including models of neuropathic and chronic inflammatory pain. Moreover, P2X7R activity, by regulating the release of pro-inflammatory cytokines, may be involved in the pathophysiology of neuropsychiatric disorders. The P2X7R may thus represent a critical communication link between the nervous and immune systems, while providing a target for therapeutic exploitation. In this review we discuss current biology and pharmacology of the P2X7R, as well as insights into the role for this receptor in neurological/psychiatric diseases.


Subject(s)
Epilepsy/metabolism , Neuralgia/metabolism , Neuroblastoma/metabolism , Neurodegenerative Diseases/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/physiology , Animals , Humans , Ion Channels , Mice , Permeability , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism
2.
Curr Pharm Des ; 17(25): 2704-18, 2011.
Article in English | MEDLINE | ID: mdl-21728978

ABSTRACT

Neurotrophic factors were originally identified based on their ability to prevent naturally occurring cell death in the developing nervous system. Many of these proteins also promote survival after injury or protect neurons in toxin-disease models in animals. In addition to neuroprotective effects, these factors exert trophic effects on neurons, stimulating increases in neuronal metabolism, cell size, and process outgrowth. These properties underlie expectations for neurorestoration, in which growth of new axons and synapses could lead to functional improvement, which is of great interest for those patients who are already significantly disabled by disease. Preclinical and clinical data suggest that subcutaneous or intravenous administration of neurotrophic factors may be effective for the treatment of peripheral nervous system diseases. However, even though these proteins are natural products, they do present specific problems when used as therapeutic agents. They cannot be given orally, present uncertain pharmacokinetic behavior, and large-scale production is labor and cost-intensive. Neurotrophic factor treatment of central nervous system diseases presents an even more complex scenario, since they are not able to cross the blood-brain barrier and must be given intracerebrally. Although there is an active search for alternative delivery strategies, for central nervous system diseases in particular the advantages of small molecule mimetics over proteins are evident. Small organic molecules can be modified to penetrate freely into the brain parenchyma and can be designed for oral administration. There are several possible approaches for replacing neurotrophic proteins with small molecule mimetics. For therapeutic use in the peripheral nervous system, neurotrophic proteins could be replaced by active peptide fragments with receptor binding properties similar to the full-length protein, but improved pharmacokinetic properties and lower production costs. In principle, it should be possible to replace the entire protein or fully active peptide fragment by a non-peptidic molecule binding to the same receptor site. It may be possible to regulate neurotrophic factor receptor activity by allosterically-acting molecules which influence the functional efficacy of the receptors. Other strategies include intracellular effector-targeting approaches, which are based on knowledge of signaling pathways involved in neuronal cell survival and demise, and which can be agonized or antagonized to promote neuroprotection. This chapter will begin with a brief overview on the biology neurotrophic proteins, followed with a description of strategies taken towards the development of small molecule mimetics for neurotrophic factors and the emerging drug candidates. The latter will encompass both receptor-directed as well as intracellular signalling approaches.


Subject(s)
Nerve Growth Factors/metabolism , Peptides/pharmacology , Receptors, Nerve Growth Factor/metabolism , Animals , Drug Design , Humans , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/pharmacology , Nervous System Diseases/drug therapy , Nervous System Diseases/physiopathology , Peptides/administration & dosage , Peptides/pharmacokinetics , Signal Transduction/drug effects
3.
J Neurochem ; 95(3): 715-23, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16104849

ABSTRACT

Amyloid beta-peptide (Abeta) is the main component of senile plaques which characterize Alzheimer's disease and may induce neuronal death through mechanisms which include oxidative stress. To date, the signalling pathways linking oxidant stress, a component of several neurodegenerative diseases, to cell death in the CNS are poorly understood. Melastatin-like transient receptor potential 2 (TRPM2) is a Ca(2+)-permeant non-selective cation channel, which responds to increases in oxidative stress levels in the cell and is activated by oxidants such as hydrogen peroxide. We demonstrate here that Abeta and hydrogen peroxide both induce death in cultured rat striatal cells which express TRPM2 endogenously. Transfection with a splice variant that acts as a dominant negative blocker of TRPM2 function (TRPM2-S) inhibited both hydrogen peroxide- and Abeta-induced increases in intracellular-free Ca(2+) and cell death. Functional inhibition of TRPM2 activation by the poly(ADP-ribose)polymerase inhibitor SB-750139, a modulator of intracellular pathways activating TRPM2, attenuated hydrogen peroxide- and Abeta-induced cell death. Furthermore, a small interfering RNA which targets TRPM2, reduced TRPM2 mRNA levels and the toxicity induced by hydrogen peroxide and Abeta. These data demonstrate that activation of TRPM2, functionally expressed in primary cultures of rat striatum, contributes to Abeta- and oxidative stress-induced striatal cell death.


Subject(s)
Amyloid beta-Peptides/toxicity , Clusterin/metabolism , Hydrogen Peroxide/toxicity , Neurons/metabolism , Oxidants/toxicity , Peptide Fragments/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Calcium/metabolism , Cell Death/drug effects , Cells, Cultured , Clusterin/genetics , Corpus Striatum/cytology , Neurons/cytology , Neurons/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
4.
Curr Drug Targets CNS Neurol Disord ; 2(5): 279-91, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529360

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1) protects the genome by functioning in the DNA damage surveillance network. In response to stresses that are toxic to the genome, PARP-1 activity increases substantially, an event that appears crucial for maintaining genomic integrity. Massive PARP-1 activation, however, can deplete the cell of NAD(+) and ATP, ultimately leading to energy failure and cell death. The discovery that cell death may be suppressed by PARP inhibitors or by deletion of the parp-1 gene has prompted a great deal of interest in the process of poly(ADP-ribosyl)ation. Suppression of PARP-1 is capable of protecting against cerebral and cardiac ischemia, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism, traumatic spinal cord injury, and streptozotocin-induced diabetes. The secondary damage of initially surviving neurons in brain stroke accounts for most of the volume of the infarcted area and the subsequent loss of brain function. Microglial migration is strongly controlled in living brain tissue by expression of the integrin CD11a, which is regulated in turn by PARP-1, proposing that PARP-1 downregulation may therefore be a promising strategy in protecting neurons from this secondary damage, as well. As PARP-1 is now recognised as playing a role also in the regulation of gene transcription, this further increases the intricacy of poly(ADP-ribosyl)ation in the control of cell homeostasis and challenges the notion that energy collapse is the sole mechanism by which poly(ADP-ribose) formation contributes to cell death. PARP(s) might regulate cell fate as essential modulators of death and survival transcriptional programs with relation to NF-kappaB and p53, proposing that inhibitors of poly(ADP-ribosyl)ation could therefore prevent the deleterious consequences of neuroinflammation by reducing NF-kappaB activity.


Subject(s)
Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/enzymology , Drug Delivery Systems/methods , Neuroprotective Agents/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Acute Disease , Animals , Humans
5.
Neuropharmacology ; 45(5): 623-36, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12941376

ABSTRACT

Corticotropin-releasing factor (CRF) receptors are members of the superfamily of G-protein coupled receptors that utilise adenylate cyclase and subsequent production of cAMP for signal transduction in many tissues. Activation of cAMP-dependent pathways, through elevation of intracellular cAMP levels is known to promote survival of a large variety of central and peripheral neuronal populations. Utilising cultured primary rat central nervous system neurons, we show that stimulation of endogenous cAMP signalling pathways by forskolin confers neuroprotection, whilst inhibition of this pathway triggers neuronal death. CRF and the related CRF family peptides urotensin I, urocortin, and sauvagine, which also induced cAMP production, prevented the apoptotic death of cerebellar granule neurons triggered by inhibition of phosphatidylinositol kinase-3 pathway activity with LY294002. These effects were negated by the highly selective CRF-R1 antagonist CP154,526. CRF even conferred neuroprotection when its application was delayed by up to 8 h following LY294002 addition. The CRF peptides also protected cortical and hippocampal neurons against death induced by beta-amyloid peptide (1-42), in a CRF-R1 dependent manner. In separate experiments, LY294002 reduced neuronal protein kinase B activity while increasing glycogen synthase kinase-3, whilst CRF (and related peptides) promoted phosphorylation of glycogen synthase kinase-3 without protein kinase B activation. Taken together, these results suggest that the neuroprotective activity of CRF may involve cAMP-dependent phosphorylation of glycogen synthase kinase-3.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP/analogs & derivatives , Neurons/drug effects , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases , Receptors, Corticotropin-Releasing Hormone/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Amphibian Proteins , Amyloid beta-Peptides/pharmacology , Animals , Animals, Newborn , Apoptosis , Blotting, Western/methods , Cell Survival/drug effects , Cells, Cultured , Cerebellum , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Chromatin/metabolism , Chromones/pharmacology , Colforsin/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Glycogen Synthase Kinase 3/metabolism , Hippocampus/drug effects , Hippocampus/physiology , In Situ Nick-End Labeling/methods , Inhibitory Concentration 50 , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Peptide Fragments/pharmacology , Peptide Hormones , Peptides/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Thionucleotides/pharmacology , Time Factors , Urocortins , Urotensins/pharmacology , Vasodilator Agents/pharmacology
6.
Brain Res Mol Brain Res ; 97(2): 177-85, 2001 Dec 30.
Article in English | MEDLINE | ID: mdl-11750074

ABSTRACT

Nerve growth factor (NGF), a target-derived factor for survival and maintenance of peripheral and central neurons, has been implicated in inflammatory processes. Mast cells are the principal effector cells in IgE-dependent hypersensitivity reactions, and also play a role in diseases characterised by inflammation, including those of the nervous system like multiple sclerosis. Mast cells are capable of synthesising and responding to NGF, although the occurrence of other members of the NGF family of neurotrophins and their protein forms have not been described. Immunoblot analysis with highly selective neurotrophin antibodies has now been used to show that rat peritoneal mast cells express a higher molecular weight form (73 kDa) of NGF, but not the monomeric (13 kDa) NGF polypeptide. Mast cells also expressed 73 kDa forms of neurotrophin-4 and neurotrophin-3; brain-derived neurotrophic factor was not detected. Medium conditioned by degranulating peritoneal mast cells contained similar high molecular weight forms of NGF and neurotrophin-4 on Western blots, but no neurotrophin-3. Mast cell-derived neurotrophin immunoreactivities were inhibited by the respective peptide antigen, further demonstrating the specificity of the mast cell-derived neurotrophic protein. Mast cell-released proteins supported the survival of cultured chicken embryonic neural crest- and placode-derived sensory neurons; neurotrophic activities were inhibited by neutralising antibodies for NGF and neurotrophin-4, respectively. High molecular isoforms of neurotrophins have been reported to occur in experimental colitis and in the inflamed gut of patients with Crohn's disease and ulcerative colitis, tissue sites rich in mast cells. The data suggest an important role for neurotrophins in the pathophysiology of inflammatory disease.


Subject(s)
Mast Cells/metabolism , Nerve Growth Factors/biosynthesis , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/chemistry , Cell Degranulation , Cell Survival , Male , Mast Cells/chemistry , Molecular Weight , Nerve Growth Factor/analysis , Nerve Growth Factor/chemistry , Nerve Growth Factors/analysis , Nerve Growth Factors/chemistry , Neurons, Afferent/chemistry , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Neurotrophin 3/analysis , Neurotrophin 3/biosynthesis , Neurotrophin 3/chemistry , Nodose Ganglion/cytology , Peritoneal Cavity , Rats , Rats, Sprague-Dawley , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Serotonin/metabolism , Tritium
7.
Prog Neurobiol ; 65(6): 593-608, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11728645

ABSTRACT

During development of the nervous system, neurons extend axons over considerable distances in a highly stereospecific fashion in order to innervate their targets in an appropriate manner. This involves the recognition, by the axonal growth cone, of guidance cues that determine the pathway taken by the axons. These guidance cues can act to promote and/or repel growth cone advance, and they can act either locally or at a distance from their place of synthesis. The directed growth of axons is partly governed by cell adhesion molecules (CAMs) on the neuronal growth cone that bind to CAMs on the surface of other axons or non-neuronal cells. In vitro assays have established the importance of the CAMs (N-CAM, N-cadherin and the L1 glycoprotein) in promoting axonal growth over cells, such as Schwann cells, astrocytes and muscle cells. Strong evidence now exists implicating the fibroblast growth factor receptor tyrosine kinase as the primary signal transduction molecule in the CAM pathway. Cell adhesion molecules are important constituents of synapses, and CAMs appear to play important and diverse roles in regulating synaptic plasticity associated with learning and memory. Negative extracellular signals which physically direct neurite growth have also been described. The latter include the neuronal growth inhibitory proteins Nogo and myelin-associated glycoprotein, as well as the growth cone collapsing Semaphorins/neuropilins. Although less well characterised, evidence is now beginning to emerge describing a role for Rho kinase-mediated signalling in inhibition of neurite outgrowth. This review focuses on some of the major themes and ideas associated with this fast-moving field of neuroscience.


Subject(s)
Cell Adhesion Molecules/physiology , Growth Inhibitors/physiology , Nervous System/growth & development , Neurons/physiology , Signal Transduction/physiology , Animals , Axons/diagnostic imaging , Axons/physiology , Brain Chemistry/physiology , Humans , Neuronal Plasticity/physiology , Neurons/cytology , Ultrasonography
8.
J Biol Chem ; 276(47): 43879-86, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11571292

ABSTRACT

In this study, we show that the neurite outgrowth response stimulated by N-cadherin is inhibited by a recently developed and highly specific fibroblast growth factor receptor (FGFR) antagonist. To test whether the N-cadherin response also requires FGF function, we developed peptide mimetics of the receptor binding sites on FGFs. Most mimetics inhibit the neurite outgrowth response stimulated by FGF in the absence of any effect on the N-cadherin response. The exceptions to this result were two mimetics of a short FGF1 sequence, which has been shown to interact with the region of the FGFR containing the histidine-alanine-valine motif. These peptides inhibited FGF and N-cadherin responses with similar efficacy. The histidine-alanine-valine region of the FGFR has previously been implicated in the N-cadherin response, and a candidate interaction site has been identified in extracellular domain 4 of N-cadherin. We now show that antibodies directed to this site on N-cadherin inhibit the neurite outgrowth response stimulated by N-cadherin, and peptide mimetics of the site inhibit N-cadherin and FGF responses. Thus, we can conclude that N-cadherin contains a novel motility motif in extracellular domain 4, and that peptide mimetics of this motif can interact with the FGFR.


Subject(s)
Axons , Cadherins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cadherins/chemistry , Cell Line , Humans , Molecular Mimicry , Molecular Sequence Data
10.
Ann N Y Acad Sci ; 939: 11-22, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11462762

ABSTRACT

Perturbation of normal survival mechanisms may play a role in a large number of disease processes. Glutamate neurotoxicity, particularly when mediated by the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors, has been hypothesized to underlie several types of acute brain injury, including stroke. Several neurological insults linked to excessive release of glutamate and neuronal death result in tyrosine kinase activation, including p44/42 mitogen activated protein (MAP) kinase. To further explore a role for MAP kinase activation in excitotoxicity, we used a novel tissue culture model to induce neurotoxicity. Removal of the endogenous blockade by Mg2+ of the NMDA receptor in cultured hippocampal neurons triggers a self perpetuating cycle of excitotoxicity, which has relatively slow onset, and is critically dependent on NMDA receptors and activation of voltage gated Na+ channels. These injury conditions led to a rapid phosphorylation of p44/42 that was blocked by MAP kinase kinase (MEK) inhibitors. MEK inhibition was associated with protection against synaptically mediated excitotoxicity. Interestingly, hippocampal neurons preconditioned by a sublethal exposure to Mg(2+)-free conditions were rendered resistant to injury induced by a subsequently longer exposure to this insult; the preconditioning effect was MAP kinase dependent. The MAP kinase signaling pathway can also promote polypeptide growth factor mediated neuronal survival. MAP kinase regulated pathways may act to promote survival or death, depending upon the cellular context in which they are activated.


Subject(s)
Brain/metabolism , Cell Death/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Animals , Brain/blood supply , Brain/drug effects , Cell Death/drug effects , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Ischemic Preconditioning , MAP Kinase Signaling System/drug effects , Magnesium/pharmacology , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/drug effects , Nerve Growth Factors/metabolism , Neurons/drug effects , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
11.
J Neurochem ; 77(1): 94-102, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11279265

ABSTRACT

The phosphatidylinositol 3-kinase (PI 3-kinase)/protein kinase B (PKB; also known as Akt) signalling pathway is recognized as playing a central role in the survival of diverse cell types. Glycogen synthase kinase-3 (GSK-3) is a ubiquitously expressed serine/threonine protein kinase that is one of several known substrates of PKB. PKB phosphorylates GSK-3 in response to insulin and growth factors, which inhibits GSK-3 activity and leads to the modulation of multiple GSK-3 regulated cellular processes. We show that the novel potent and selective small-molecule inhibitors of GSK-3; SB-415286 and SB-216763, protect both central and peripheral nervous system neurones in culture from death induced by reduced PI 3-kinase pathway activity. The inhibition of neuronal death mediated by these compounds correlated with inhibition of GSK-3 activity and modulation of GSK-3 substrates tau and beta-catenin. Thus, in addition to the previously assigned roles of GSK-3, our data provide clear pharmacological and biochemical evidence that selective inhibition of the endogenous pool of GSK-3 activity in primary neurones is sufficient to prevent death, implicating GSK-3 as a physiologically relevant principal regulatory target of the PI 3-kinase/PKB neuronal survival pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases , Trans-Activators , Aminophenols/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Chromones/pharmacology , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Indoles/pharmacology , Maleimides/pharmacology , Morpholines/pharmacology , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Substrate Specificity , beta Catenin , tau Proteins/metabolism
12.
J Neurochem ; 76(1): 47-55, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145977

ABSTRACT

Excessive glutamatergic neurotransmission, particularly when mediated by the N:-methyl-D-aspartate (NMDA) subtype of glutamate receptor, is thought to underlie neuronal death in a number of neurological disorders. Histamine has been reported to potentiate NMDA receptor-mediated events under a variety of conditions. In the present study we have utilized primary hippocampal neurone cultures to investigate the effect of mast cell-derived, as well as exogenously applied, histamine on neurotoxicity evoked by excessive synaptic activity. Exposure of mature cultures for 15 min to an Mg(2+)-free/glycine-containing buffer to trigger synaptic transmission through NMDA receptors, caused a 30-35% neuronal loss over 24 h. When co-cultured with hippocampal neurones, activated mast cells increased excitotoxic injury to 60%, an effect that was abolished in the presence of histaminase. Similarly, addition of histamine during magnesium deprivation produced a concentration-dependent potentiation (+ 60%; EC(50) : 5 microM) of neuronal death which was inhibited by sodium channel blockers and NMDA receptor antagonists, although this effect did not involve known histamine receptors. The histamine effect was further potentiated by acidification of the culture medium. Cultures 'preconditioned' by sublethal (5 min) Mg(2+) deprivation exhibited less neuronal death than controls when exposed to a more severe insult. NMDA receptor activation and the extracellular regulated kinase cascade were required for preconditioning neuroprotection. The finding that histamine potentiates NMDA receptor-mediated excitotoxicity may have important implications for our understanding of conditions where enhanced glutamatergic neurotransmission is observed in conjunction with tissue acidification, such as cerebral ischaemia and epilepsy.


Subject(s)
Hippocampus/metabolism , Histamine/metabolism , Mast Cells/metabolism , Neurons/metabolism , Synapses/metabolism , Age Factors , Animals , Binding Sites/physiology , Cell Count , Cells, Cultured , Coculture Techniques , Drug Synergism , Drug Tolerance , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Histamine/pharmacology , Ischemic Preconditioning , Magnesium/metabolism , Mast Cells/cytology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Histamine/metabolism , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology
13.
FASEB J ; 15(1): 164-170, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11149904

ABSTRACT

Unilateral injection into the right substantia nigra of the catecholaminergic neurotoxin 6-hydroxydopamine (6-OHDA) produces extensive loss of dopaminergic cells ('hemi-parkinsonian rat'). The pineal hormone melatonin, which is a potent antioxidant against different reactive oxygen species and has been reported to be neuroprotective in vivo and in vitro, was evaluated for potential anti-Parkinson effects in this model. Imbalance in dopaminergic innervation between the striata produced by intranigral administration of 6-OHDA results in a postural asymmetry causing rotation away from the nonlesioned side. Melatonin given systemically prevented apomorphine-induced circling behavior in 6-OHDA-lesioned rats. Reduced activity of mitochondrial oxidative phosphorylation enzymes has been suggested in some neurodegenerative diseases; in particular, selective decrease in complex I activity is observed in the substantia nigra of Parkinson's disease patients. Analysis of mitochondrial oxidative phosphorylation enzyme activities in nigral tissue from 6-OHDA-lesioned rats by a novel BN-PAGE histochemical procedure revealed a clear loss of complex I activity, which was protected against in melatonin-treated animals. A good correlation between behavioral parameters and enzymatic (complex I) analysis was observed independent of melatonin administration. A deficit in mitochondrial complex I could conceivably contribute to cell death in parkinsonism via free radical mechanisms, both directly via reactive oxygen species production and by decreased ATP synthesis and energy failure. Melatonin may have potential utility in the treatment of neurodegenerative disorders where oxidative stress is a participant.


Subject(s)
Melatonin/pharmacology , Mitochondria/enzymology , Neuroprotective Agents/pharmacology , Oxidative Phosphorylation/drug effects , Oxidopamine/antagonists & inhibitors , Parkinson Disease, Secondary/chemically induced , Parkinson Disease/drug therapy , Adenosine Triphosphatases/metabolism , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Disease Models, Animal , Electron Transport Complex IV/metabolism , Electrophoresis, Polyacrylamide Gel , Male , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , NAD/metabolism , Oxidopamine/administration & dosage , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Substantia Nigra/metabolism , Substantia Nigra/pathology
14.
Mol Neurobiol ; 24(1-3): 183-99, 2001.
Article in English | MEDLINE | ID: mdl-11831552

ABSTRACT

Nerve growth factor (NGF) is widely recognized as a target-derived factor responsible for the survival and maintenance of the phenotype of specific subsets of peripheral neurons and basal forebrain cholinergic nuclei during development and maturation. Other NGF-responsive cells are now known to belong to the hemopoietic-immune system and to populations in the brain involved in neuroendocrine functions. The concentration of NGF is elevated in a number of inflammatory and autoimmune states in conjunction with increased accumulation of mast cells. Mast cells and NGF appear to be involved in neuroimmune interactions and tissue inflammation. Mast cells themselves are capable of producing and responding to NGF, suggesting that alterations in mast cell behavior may trigger maladaptive neuroimmune tissue responses, including those of an autoimmune nature. Moreover, NGF exerts a modulatory role on sensory nociceptive nerve physiology in the adult, and appears to correlate with hyperalgesic phenomena occurring in tissue inflammation. NGF can thus be viewed as a multifactorial modulator of neuroimmune-endocrine functions.


Subject(s)
Immune System/physiology , Nerve Growth Factor/physiology , Neurons/physiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Humans , Mast Cells/physiology , Models, Neurological
15.
J Neurochem ; 75(4): 1520-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10987832

ABSTRACT

Basic fibroblast growth factor (FGF-2) promotes survival and/or neurite outgrowth from a variety of neurons in cell culture and regenerative processes in vivo. FGFs exert their effects by activating cell surface receptor tyrosine kinases. FGF receptor (FGFR) inhibitors have not been characterized on neuronal cell behaviors to date. In the present study, we show that the FGFR1 inhibitor PD 173074 potently and selectively antagonized the neurotrophic and neurotropic actions of FGF-2. Nanomolar concentrations of PD 173074 prevented FGF-2, but not insulin-like growth factor-1, support of cerebellar granule neuron survival under conditions of serum/K(+) deprivation; another FGF-2 inhibitor, SU 5402, was effective only at a 1,000-fold greater concentration. Neither PD 173074 nor SU 5402, at 100 times their IC(50) values, interfered with the survival of dorsal root ganglion neurons promoted by nerve growth factor, ciliary neurotrophic factor, or glial cell line-derived neurotrophic factor. PD 173074 and SU 5402 displayed 1,000-fold differential IC(50) values for inhibition of FGF-2-stimulated neurite outgrowth in PC12 cells and in granule neurons, and FGF-2-induced mitogen-activated protein kinase (p44/42) phosphorylation. The two inhibitors failed to disturb downstream signalling stimuli of FGF-2. PD 173074 represents a valuable tool for dissecting the role of FGF-2 in normal and pathological nervous system function without compromising the actions of other neurotrophic factors.


Subject(s)
Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Nerve Growth Factors , Neurons/drug effects , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/pharmacology , Dose-Response Relationship, Drug , Glial Cell Line-Derived Neurotrophic Factor , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/pharmacology , Neurites/drug effects , Neurons/cytology , PC12 Cells , Phosphorylation/drug effects , Protein-Tyrosine Kinases/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1 , Sensitivity and Specificity
16.
Brain Res ; 862(1-2): 83-9, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10799672

ABSTRACT

Glutathione (GSH) is a key component of the cellular defence cascade against injury caused by reactive oxygen species. Kainic acid (KA) is a potent central nervous system excitotoxin. KA-elicited neuronal death may result from the generation of ROS. The present study was undertaken to characterize the role of GSH in KA-induced neurotoxicity. Cultures of cerebellar granule neurons were prepared from 8-day-old rats, and used at 8, 14 and 20 days in vitro (DIV). Granule neurons displayed a developmental increase in their sensitivity to KA injury, as quantified by an ELISA-based assay with the tetrazolium salt MTT. At DIV 14 and 20, a 30-min challenge with KA (500 microM) reduced cell viability by 45% after 24 h, significantly greater (P<0.01) than the 22% cell loss with DIV 8 cultures. Moreover acute (30 min) KA exposure concentration-dependently reduced intracellular GSH and enhanced reactive oxygen species generation (evaluated by 2', 7'-dichlorofluorescein diacetate). In comparison to control, KA (500 microM) lowered GSH levels in DIV 8 granule neurons by 16% (P=0. 0388), and by 36% (P=0.0001) in both DIV 14 and DIV 20 neurons, after 30 min. Preincubation of granule neurons with the membrane permeant GSH delivery agent, GSH ethyl ester (5 mM), for 30 min significantly increased intracellular GSH content. Importantly, GSH ethyl ester reduced the toxic effects of KA, becoming significant at 1 mM (P=0.007 vs. KA-treated group), and was maximal at >/=2.5 mM (P<0.0001). GSH ethyl ester displayed a similar dose-dependence in its ability to counteract KA-induced depletion of cellular GSH. The data strengthen the notion that cellular GSH levels have a fundamental role in KA-induced neurotoxicity.


Subject(s)
Cerebellum/cytology , Excitatory Amino Acid Agonists/toxicity , Glutathione/analysis , Kainic Acid/toxicity , Nerve Degeneration/chemically induced , Neurons/chemistry , Animals , Cell Survival/drug effects , Cerebellum/chemistry , Cerebellum/metabolism , Fluoresceins , Glutamic Acid/toxicity , Glutathione/analogs & derivatives , Glutathione/pharmacology , N-Methylaspartate/toxicity , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Radiation-Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
17.
J NeuroAIDS ; 2(2): 1-13, 1999.
Article in English | MEDLINE | ID: mdl-16873189

ABSTRACT

gp120 induction of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) was studied in cultures of purified astrocytes. Incubation of pure mouse cortical astrocytes with gp120 IIIB induced the expression of TNF-alpha mRNA, assessed by in situ hybridization. Anti- TNF-alpha immunocytochemical staining of gp120 IIIB stimulated astrocytes indicated the presence of TNF-alpha. gp120 IIIB treatment also stimulated secretion of bioactive TNF-alpha from astrocytes, which was prevented by inhibitors of transcription and translation. Hippocampal and cerebellar astrocytes displayed similar behaviors. Further, gp120 displayed cytotoxicity for astrocytes that depended on macromolecular synthesis. The data are the first to show gp120 IIIB induction of de novo TNF-alpha production by pure astrocytes. Because TNF-alpha exerts a wide array of effects in the brain of infected individuals and has HIV-1 inducing activity as well, induction of this cytokine by gp120 IIIB in astrocytes may contribute importantly to the pathogenesis of AIDS dementia complex. Since TNF-alpha can stimulate astrocyte reactivity and proliferation by an autocrine mechanism, the extent of the gp120 effect could conceivably increase with HIV-1 disease progression in a self-amplifying loop, involving other cell types, thus favoring both virus persistence and a chronic disease state.


Subject(s)
Astrocytes/virology , HIV Envelope Protein gp120/pharmacology , HIV-1/physiology , Tumor Necrosis Factor-alpha/genetics , Animals , Astrocytes/physiology , Cerebral Cortex/virology , Hippocampus/virology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics
18.
Adv Exp Med Biol ; 467: 207-15, 1999.
Article in English | MEDLINE | ID: mdl-10721058

ABSTRACT

The physiological roles of the pineal hormone melatonin are still not completely clarified. Recently it has been shown that melatonin is a potent, endogenous scavenger of reactive oxygen species suggesting that it might interfere with neurodegenerative processing involving free-radical formation and excitatory aminoacid release. These neuroprotective effects of melatonin may result, at least in part, from a sparing of glutathione reductase, which is decreased following administration of the neurotoxic agent kainate (KA) in rats. Moreover, KA causes a rapid decrease in glutathione (GSH) content of cultured cerebellar granule neurons but not in astrocytes. These cell types both express functional KA receptors, but only the former is sensitive to reactive oxygen species-dependent KA injury. Melatonin counteracts the changes in GSH, induced by KA, in cultured cerebellar granule neurons.


Subject(s)
Brain/physiology , Melatonin/pharmacology , Melatonin/physiology , Neuroprotective Agents/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Brain/drug effects , Free Radical Scavengers/pharmacology , Glutathione/metabolism , Glutathione Reductase/metabolism , Humans , Kainic Acid/toxicity , Neurons/drug effects , Neurons/physiology , Rats
19.
Ann N Y Acad Sci ; 890: 107-18, 1999.
Article in English | MEDLINE | ID: mdl-10668417

ABSTRACT

The brain consumes large quantities of oxygen relative to its contribution to total body mass. This, together with its paucity of oxidative defense mechanisms, places this organ at risk for damage mediated by reactive oxygen species. The pineal secretory product melatonin possesses broad-spectrum free radical scavenging and antioxidant activities, and prevents kainic acid-induced neuronal lesions, glutathione depletion, and reactive oxygen species-mediated apoptotic nerve cell death. Melatonin's action is thought to involve electron donation to directly detoxify free radicals such as the highly toxic hydroxyl radical, which is a probable end-product of the reaction between NO. and peroxynitrite. Moreover, melatonin limits NO.-induced lipid peroxidation, inhibits cerebellar NO. synthase, scavenges peroxynitrite, and alters the activities of enzymes that improve the total antioxidative defense capacity of the organism. Melatonin function as a free radical scavenger and antioxidant is likely facilitated by the ease with which it crosses morphophysiological barriers, e.g., the blood-brain barrier, and enters cells and subcellular compartments. Pinealectomy, which eliminates the nighttime rise in circulating and tissue melatonin levels, worsens both reactive oxygen species-mediated tissue damage and brain damage after focal cerebral ischemia and excitotoxic seizures. That melatonin protects against hippocampal neurodegeneration linked to excitatory synaptic transmission is fully consistent with the last study. Conceivably, the decreased melatonin secretion that is documented to accompany the aging process may be exaggerated in populations with dementia.


Subject(s)
Antioxidants/pharmacology , Melatonin/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Excitatory Amino Acid Agonists/pharmacology , Glutathione/drug effects , Glutathione/metabolism , Kainic Acid/pharmacology , Melatonin/metabolism , Neurotoxins/pharmacology , Oxidative Stress/physiology , Rats
20.
Mol Cell Neurosci ; 12(4-5): 179-93, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9828084

ABSTRACT

Over the past several years, neurotrophic factors-a description generally applied to naturally occurring polypeptides that support the development and survival of neurons-have made considerable progress from the laboratory into the clinic. Evidence from preclinical and clinical studies indicates that it may be possible to use neurotrophic factors to prevent, slow the progression of, or even reverse the effects of a number of neurodegenerative diseases and other types of insults in both the central nervous system (CNS) and the peripheral nervous system. Initially, investigations focused on recombinant neurotrophic proteins that are identical or highly homologous to the natural human sequence. Given the difficulties inherent with a protein therapeutic approach to treating nervous system disorders, especially those of the CNS, increasing attention has now turned to the development of alternative strategies and, in particular, small molecule mimetics. Regulation of the transcription of neurotrophic factors may provide a means of manipulating endogenous factor production; gene therapy may also allow for the circumvention of exogenous neurotrophic factor administration. The problem of transport across the blood-brain barrier may be overcome by developing small-molecule mimetics that maintain the neurotrophic activity of the protein while having improved pharmacokinetic and disposition characteristics. Components of neurotrophic factor signal transduction pathways may provide additional targets for novel drugs that can induce or modulate the responses normally activated by the binding of the neurotrophic factor to its receptor. This review focusses on some of the major themes and lines of mechanistic and therapeutic advances in this fast-moving field of neuroscience.


Subject(s)
Nerve Degeneration/drug therapy , Nerve Growth Factors/physiology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/physiopathology , Drug Design , Humans , Nerve Degeneration/physiopathology , Nerve Growth Factors/chemistry , Nerve Growth Factors/therapeutic use , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/physiopathology , Receptors, Nerve Growth Factor/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL