Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 37(7): 1508-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22528836

ABSTRACT

Activation of RAGE (receptor for advanced glycation endproducts) and of its subtypes may play a role in neuronal damage and neuroinflammation associated with brain ischemia, though the underlying mechanisms remain unclear. In this study, we have examined by Western blotting the expression of RAGE isoforms in the cerebral cortex and striatum of Wistar rats subjected to transient (1 or 2 h) middle cerebral artery occlusion (tMCAo). The findings show that the full-length RAGE (~50 kDa) and its isoforms in the 26-43 kDa range are significantly decreased in the ischemic cortex, but not in the striatum, after 1 and 2 h tMCAo when compared to the sham group. By contrast, in the striatum, ischemia-reperfusion injury caused a significant increase of full-length RAGE and its isoforms in the 72-100 kDa range. We also investigated the soluble form of RAGE, which was significantly decreased in the plasma of rats subjected to transient or permanent MCAo. In conclusion, the present data demonstrate that regional brain expression of RAGE is differentially affected by tMCAo in rat. These modifications are accompanied by a decrease in the plasma levels of soluble RAGE, thereby suggesting a potential role for soluble RAGE as a peripheral biomarker of focal ischemia.


Subject(s)
Ischemic Attack, Transient/metabolism , Protein Isoforms/metabolism , Receptors, Immunologic/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Ischemic Attack, Transient/blood , Male , Protein Isoforms/blood , Rats , Rats, Wistar/blood , Receptor for Advanced Glycation End Products
2.
Int Rev Neurobiol ; 82: 149-69, 2007.
Article in English | MEDLINE | ID: mdl-17678960

ABSTRACT

Abnormal expression of matrix metalloproteinases (MMPs) has been implicated in the pathophysiology of neuroinflammatory processes that accompany most central nervous system disease. In particular, early upregulation of the gelatinases MMP-2 and MMP-9 has been shown to contribute to disruption of the blood-brain barrier and to death of neurons in ischemic stroke. In situ zymography reveals a significant increase in gelatinolytic MMPs activity in the ischemic brain hemisphere after 2-h middle cerebral artery occlusion (MCAo) followed by 2-h reperfusion in rat. Accordingly, gel zymography demonstrates that expression and activity of MMP-2 and MMP-9 are enhanced in cortex and striatum ipsilateral to the ischemic insult. The latter effect appears to be instrumental for development of delayed brain damage since administration of a broad spectrum, highly specific MMPs inhibitor, GM6001, but not by its negative control, results in a significant (50%) reduction in ischemic brain volume. Increased gelatinase activity in the ischemic cortex coincides with elevation (166% vs sham) of mature interleukin-1beta (IL-1beta) after 2-h reperfusion and this does not appear to implicate a caspase-1-dependent processing of pro(31kDa)-IL-1beta to yield mature (17kDa) IL-1beta. More importantly, when administered at a neuroprotective dose GM6001 abolishes the early IL-1beta increase in the ischemic cortex and reduces the cleavage of the cytokine proform supporting the deduction that MMPs may initiate IL-1beta processing. In conclusion, development of tissue damage that follows transient ischemia implicates a crucial interplay between MMPs and mediators of neuroinflammation (e.g., IL-1beta), and this further underscores the therapeutic potential of MMPs inhibitors in the treatment of stroke.


Subject(s)
Brain Ischemia/metabolism , Inflammation Mediators/metabolism , Matrix Metalloproteinases/biosynthesis , Reperfusion Injury/enzymology , Animals , Blotting, Western , Brain Ischemia/pathology , Caspase 1/metabolism , Fluorometry , Gelatin/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Interleukin-1beta/biosynthesis , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Rats , Rats, Wistar , Up-Regulation/physiology
3.
Int Rev Neurobiol ; 82: 357-72, 2007.
Article in English | MEDLINE | ID: mdl-17678971

ABSTRACT

Neuroprotection exerted by 17beta-estradiol (17beta-E(2)) has been widely investigated in animal models of acute cerebral ischemia. Estrogens interact with intracellular receptors (ERalpha and ERbeta) to modulate the transcription of target genes, including those implicated in neuronal survival. Neuroprotection may also occur via interaction with ER-like membrane receptors mediating rapid, non-genomic, actions or via receptor-independent mechanisms. There is also evidence that blockade of inflammatory factors may represent an important mechanism involved in estrogenic neuroprotection. Here we investigate whether reduced brain damage by acute pharmacological treatment with 17beta-E(2) in male rats subjected to transient (2h) middle cerebral artery occlusion (tMCAo) involves modulation of interleukin-1beta (IL-1beta), a proinflammatory cytokine strongly implicated in the pathophysiology of ischemic stroke. Administration of 17beta-E(2) (0.2mg/kg, i.p., 1h before tMCAo) results in significant reduction of brain infarct volume, and this is reverted by the ER antagonist ICI 182,780 (0.25mg/kg, i.p.) administered 1h before 17beta-E(2). Two hours MCAo followed by 2-h reperfusion results in a significant, threefold increase of IL-1beta levels in the cortical tissue ipsilateral to the ischemic damage. Interestingly, a pretreatment with a neuroprotective dose of 17beta-E(2) attenuates the cytokine elevation and this appears to occur through ER activation. In addition, neuroprotection by 17beta-E(2) is accompanied by reduced cytochrome c translocation both in the striatum and in the cortex as revealed by Western blotting 3h after reperfusion. In conclusion, we report the original observation that neuroprotection exerted by 17beta-E(2) in a rat model of transient focal brain ischemia is accompanied by reduced cytochrome c translocation to the cytosol and involves early modulation of IL-1beta production.


Subject(s)
Estradiol/pharmacology , Infarction, Middle Cerebral Artery/prevention & control , Interleukin-1beta/physiology , Neuroprotective Agents , Animals , Blotting, Western , Brain Ischemia/pathology , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/enzymology , Enzyme-Linked Immunosorbent Assay , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Fulvestrant , Infarction, Middle Cerebral Artery/pathology , Interleukin-1beta/biosynthesis , Male , Rats , Rats, Wistar , Receptors, Estrogen/antagonists & inhibitors , Subcellular Fractions/drug effects , Subcellular Fractions/pathology
4.
Int Rev Neurobiol ; 82: 397-406, 2007.
Article in English | MEDLINE | ID: mdl-17678974

ABSTRACT

Recent studies support a role for excitotoxicity in the development of retinal ganglion cell (RGC) damage in subjects suffering from glaucoma. Coenzyme Q10 (CoQ10), an essential cofactor of the electron transport chain, has been reported to afford neuroprotection, preventing the formation of the mitochondrial permeability transition pore. Using an established animal model of retinal ischemia/reperfusion here, we show that synaptic glutamate increases at 130min from beginning of reperfusion and delayed apoptosis in the RGC layer is seen at 24h. Intraocular administration of CoQ10 minimizes glutamate increase and affords neuroprotection, suggesting that oxidative stress and energy failure might be implicated in the mechanisms of RGC death.


Subject(s)
Intraocular Pressure/physiology , Ischemia/pathology , Retinal Diseases/prevention & control , Ubiquinone/analogs & derivatives , Administration, Topical , Animals , Coenzymes/administration & dosage , Coenzymes/therapeutic use , Glutamic Acid/toxicity , In Situ Nick-End Labeling , Male , Microdialysis , Rats , Rats, Wistar , Retinal Diseases/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Ubiquinone/administration & dosage , Ubiquinone/therapeutic use
5.
Int Rev Neurobiol ; 82: 423-35, 2007.
Article in English | MEDLINE | ID: mdl-17678976

ABSTRACT

Transient focal ischemia caused by middle cerebral artery occlusion (MCAo) produces apoptotic cell death in the penumbra area. Bcl-2 is a protooncogene that plays a major antiapoptotic role, at the cellular level, by counteracting the activation of apoptosis effectors, that is, caspases. It has been suggested that nitroglycerin (NTG), a nitric oxide donor, reduces ischemia/reperfusion-induced brain damage via the inhibition of caspase activity and NMDA receptor. In this chapter, we evaluated the protective effects of NTG against cerebral damage caused by transient (2h) MCAo (tMCAo) focusing our interest on the potential effects on Bcl-2 expression. Male Wistar rats were administered intraperitoneally (i.p.) with NTG (10mg/kg) or vehicle (PEG, 1ml/kg) 20min before the induction of MCAo by intraluminal silicon-coated filament (0.37-mm diameter). Cerebral infarct volume was measured 22h after reperfusion, while cortical Bcl-2 expression was evaluated at the end of 2-h MCAo (without reperfusion) and at 5h of reperfusion. The results show significant reduction of the infarct volume in rats preinjected with NTG, as compared to the vehicle group. After 2h of occlusion, no significant difference was seen in Bcl-2 expression in the ipsilateral and contralateral cortex of either experimental groups (NTG and vehicle). However, 5h after reperfusion, a significant increase of Bcl-2 expression was detected in the damaged cortex of control rats, probably reflecting a compensatory response aiming at counteracting the cell death process; this increase was absent in the NTG-treated rats. These data, while confirming the neuroprotective effect of NTG in an in vivo ischemia/reperfusion model, seem to suggest that the drug may act by downsizing the complex chain of events underlying apoptosis activation and consequent activation of antiapoptotic responses.


Subject(s)
Brain Ischemia/complications , Genes, bcl-2/drug effects , Neuroprotective Agents , Nitroglycerin/pharmacology , Stroke/drug therapy , Vasodilator Agents/pharmacology , Animals , Blotting, Western , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Functional Laterality/physiology , Gene Expression/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Male , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Stroke/etiology , Stroke/pathology
6.
Int Rev Neurobiol ; 82: 407-21, 2007.
Article in English | MEDLINE | ID: mdl-17678975

ABSTRACT

Neuroinflammation is often associated with neurodegenerative diseases, including multiple sclerosis (MS), stroke, Alzheimer's disease, and HIV-1-associated dementia (HAD). The proinflammatory cytokine interleukin-1beta (IL-1beta) is one of the main mediators of inflammation, and IL-1beta expression in the brain is rapidly upregulated in response to acute and chronic insults. IL-1beta is synthesized as biologically inactive precursor (pro-IL-1beta), which is classically processed by caspase-1 [also known as interleukin-converting enzyme (ICE)] into the active, mature cytokine. However, caspase-1/ICE-independent mechanisms of IL-1beta processing have recently been suggested. Here we report that matrix metalloproteinases (MMPs) participate in the maturation process (cleavage and activation) of IL-1beta in an in vivo model of HIV-associated neurodegeneration based on the intracerebroventricular injection of the HIV-1 envelope glycoprotein gp120. We show that, following gp120 exposure, MMP-9 and MMP-2, but not caspase-1/ICE, are rapidly induced. Pharmacological manipulation of MMPs activity, using the broad spectrum MMPs inhibitor GM6001, reduces the increase in IL-1beta immunoreactivity and the neuronal apoptosis induced by gp120. Taken together, these findings point to a critical role for MMPs in IL-1beta increase and consequent neurotoxicity triggered by gp120 in the neocortex of rat and suggest new links between IL-1beta processing and MMP activation during the neuroinflammatory process.


Subject(s)
Apoptosis/physiology , HIV Envelope Protein gp120/toxicity , Interleukin-1beta/physiology , Matrix Metalloproteinases/physiology , Neocortex/pathology , Neurons/physiology , Animals , Caspase 1/metabolism , Caspase Inhibitors , Dipeptides/pharmacology , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Injections, Intraventricular , Interleukin-1beta/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Neocortex/drug effects , Rats , Rats, Wistar
8.
J Neurochem ; 89(5): 1293-300, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147522

ABSTRACT

Human immunodeficiency virus type-1 coat glycoprotein gp120 causes delayed apoptosis in rat brain neocortex. Here, we investigated the possible role of the endocannabinoid system in this process. It is shown that gp120 causes a time-dependent increase in the activity and immunoreactivity of the anandamide (AEA)-hydrolyzing enzyme fatty acid amide hydrolase (FAAH), paralleled by increased activity of the AEA membrane transporter and decreased endogenous levels of AEA. The AEA-synthesizing phospholipase D and the AEA-binding receptors were not affected by gp120. None of the changes induced by gp120 in the cortex were induced by bovine serum albumin, nor were they observed in the hippocampus of the same animals. Also, the activity of 5-lipoxygenase, which generates AEA derivatives able to inhibit FAAH, decreased down to approximately 25% of the control activity upon gp120 treatment, due to reduced protein level ( approximately 45%). In addition, the FAAH inhibitor methyl-arachidonoyl fluorophosphonate significantly reduced gp120-induced apoptosis in rat brain neocortex, whereas selective blockers of AEA membrane transporter or of AEA-binding receptors were ineffective. Taken together, these results suggest that gp120, by activating FAAH, decreases endogenous levels of AEA, and the latter effect seems instrumental in the execution of delayed neuronal apoptosis in the brain neocortex of rats.


Subject(s)
Apoptosis/physiology , Arachidonic Acids/metabolism , HIV Envelope Protein gp120/toxicity , Neocortex/drug effects , Neocortex/metabolism , Neurons/metabolism , Amidohydrolases/drug effects , Amidohydrolases/metabolism , Animals , Apoptosis/drug effects , Arachidonate 5-Lipoxygenase/drug effects , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acids/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Endocannabinoids , Enzyme Inhibitors/pharmacology , HIV Envelope Protein gp120/administration & dosage , Injections, Intraventricular , Male , Neocortex/pathology , Neurons/drug effects , Neurons/pathology , Organophosphonates/pharmacology , Polyunsaturated Alkamides , Rats , Rats, Wistar
9.
Basic Clin Pharmacol Toxicol ; 94(3): 132-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15052999

ABSTRACT

The epileptogenic and neurodegenerative effects of gamma-dendrotoxin, from Dendroaspis angusticeps, a specific blocker of a non-inactivating, voltage-sensitive K+ channel, were studied after focal injection into one dorsal hippocampus in rats pretreated with CGP040116, a N-methyl-D-aspartate (NMDA) receptor antagonist, and in rats bearing a monolateral surgical lesion of the Schaffer collaterals whose terminals originate from CA3 pyramids and release glutamate in the CA1 hippocampal area. Administration of 35 pmol gamma-dendrotoxin elicited in all of the treated animals (n=8) bilateral EEG discharges and damage to the hippocampal formation. Quantitation of the damage revealed significant bilateral neuronal cell loss in the CA1, CA3 and CA4 pyramidal cell layers. The lowest dose (0.35 pmol; n=4) of the toxin used did not affect EEG activity and failed to cause significant hippocampal cell loss whereas the 3.5 pmol (n=6) dose caused EEG seizures and hippocampal cell loss limited to the CA1 area. Systematic intraperitoneal administration of CGP040116 (5mg/kg given 30 min. previously) delayed the onset of EEG seizures and reduced the number of epileptogenic discharges typically observed in rats receiving an injection of gamma-dendrotoxin (35 pmol) alone. Similarly, this treatment prevented the damage inflicted to the hippocampus by the toxin and in no instance was significant neuronal loss observed. Protection against seizures and hippocampal damage was also observed by a monolateral surgical lesion to the Schaffer collaterals. In conclusion, the present data suggest that an excitotoxic, glutamate-mediated, type of mechanism underlies seizures and hippocampal damage induced by gamma-dendrotoxin in rats.


Subject(s)
2-Amino-5-phosphonovalerate/analogs & derivatives , Glutamic Acid/physiology , Hippocampus/drug effects , Neurons/pathology , Peptides/toxicity , Potassium Channel Blockers/toxicity , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Dose-Response Relationship, Drug , Elapid Venoms/toxicity , Electroencephalography , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/pathology , Hippocampus/physiopathology , Injections, Intraperitoneal , Male , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/pathology , Seizures/physiopathology
10.
J Neurochem ; 82(6): 1444-52, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12354292

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) coat glycoprotein gp120 binds to its (co)receptors and orchestrates cell entry by the direct fusion of viral and target cell membranes. Here, we modulated membrane fluidity of human neuroblastoma CHP100 cells by modulating their cholesterol content, and investigated the ability of gp120 to induce cell death in comparison with the untreated cells. We show that in normal CHP100 cells gp120 induces necrosis by: (i) increased cyclooxygenase and 5-lipoxygenase activity, and metabolites generated thereof (prostaglandin E2 and leukotriene B4, respectively); (ii) increased membrane lipoperoxidation; and (iii) increased mitochondrial uncoupling. These events were triggered by a rapid increase in intracellular calcium, and in cholesterol-depleted cells engaged CXCR4 chemokine receptors. The intracellular calcium chelator EGTA-AM protected CHP100 cells almost completely against the toxic effects of gp120. However, gp120-induced necrosis and related biochemical changes were negligible in cholesterol-enriched, and significantly enhanced in cholesterol-depleted, CHP100 cells exposed to the viral glycoprotein under the same experimental conditions. Taken together, these results suggest that membrane fluidity may control the neurotoxic effects of HIV-1 glycoprotein gp120.


Subject(s)
Cholesterol/pharmacology , HIV Envelope Protein gp120/toxicity , HIV-1 , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acid/metabolism , Calcium/metabolism , Cell Death/drug effects , Enzyme Activation/drug effects , Humans , Intracellular Fluid/metabolism , Membrane Fluidity/drug effects , Membrane Fluidity/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis , Neuroblastoma/pathology , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Binding/drug effects , Receptors, CXCR4/biosynthesis , Tumor Cells, Cultured
11.
Biochem Biophys Res Commun ; 291(2): 255-60, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11846398

ABSTRACT

Administration of tacrine (5 mg/kg ip), an anticholinesterase agent, in rats pretreated (24 h beforehand) with lithium chloride (LiCl; 12 mEq/kg ip) provides a useful experimental model to study limbic seizures and delayed hippocampal damage. Here we report Western blotting evidence demonstrating that in rat LiCl and tacrine enhance the expression of neuronal nitric oxide synthase (nNOS), but not eNOS, enzyme protein in the hippocampus during the preconvulsive period and this triggers seizures and hippocampal damage. In fact, systemic administration of 7-nitro indazole (7-NI; 50 mg/kg given ip 30 min before tacrine), a selective inhibitor of nNOS, prevented the expression of motor and electrocortical (ECoG) seizures and abolished neuronal cell death in the hippocampus. A lower dose (5 mg/kg ip) of 7-NI was ineffective. In conclusion, the present data support a role for abnormal nNOS expression in the mechanism which triggers limbic seizures and delayed excitotoxic damage in the hippocampus of rat.


Subject(s)
Hippocampus/pathology , Nitric Oxide Synthase/biosynthesis , Seizures/enzymology , Seizures/etiology , Animals , Blotting, Western , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Hippocampus/enzymology , Indazoles/pharmacology , Lithium Chloride/antagonists & inhibitors , Male , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Rats , Rats, Wistar , Seizures/pathology , Tacrine/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...