Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 179(1-2): 94-100, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16844235

ABSTRACT

We previously showed that the cytostatic drug hydroxyurea (HU) activates the hypothalamo-pituitary-adrenal (HPA) axis in intact rats, whereas it is lethal in rats with impaired HPA function. In these animals, HU toxicity is mediated by increased circulating levels of proinflammatory cytokines, whose secretion cannot be counteracted by glucocorticoids, suggesting that HPA activation blunts HU toxicity. Here we investigated the mechanisms through which HU activates the HPA axis, looking at the direct effects of the drug on the isolated hypothalamus. We found that HU significantly increases the release of arginine vasopressin but not that of corticotrophin-releasing hormone in short-term incubation experiments. The levels of arginine vasopressin are also increased in the hypothalamus and systemic circulation 2 h after the in vivo administration of the drug. Furthermore, HU increased significantly the expression of interleukin-6 and, to a lesser extent, interleukin-1beta in the hypothalamus. Interestingly, experiments with HU on primary cultures of rat microglia and astrocytes suggested that the increase in cytokine gene expression observed in hypothalamic explants is not accounted for by glial cells. Since both vasopressin and cytokines can activate the HPA axis, our present findings provide a reasonable explanation of the HPA activation elicited by HU in vivo in the rat.


Subject(s)
Arginine Vasopressin/drug effects , Cytokines/drug effects , Enzyme Inhibitors/pharmacology , Hydroxyurea/pharmacology , Hypothalamus/drug effects , Animals , Arginine Vasopressin/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Corticotropin-Releasing Hormone/drug effects , Corticotropin-Releasing Hormone/metabolism , Cytokines/biosynthesis , Gene Expression/drug effects , Gene Expression Profiling , Hypothalamo-Hypophyseal System/drug effects , Hypothalamus/metabolism , Male , Pituitary-Adrenal System/drug effects , Rats , Rats, Wistar
2.
Int J Mol Med ; 18(1): 141-5, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16786165

ABSTRACT

In this study the potential of clinically relevant alumina ceramic and metal wear particles to induce an in vitro inflammatory response was assessed in human monocytes and lymphocytes isolated from healthy donors by measuring prostaglandin E2 (PGE2) levels and mRNA expression of various pro-inflammatory cytokines. Bacterial lipopolysaccharide (LPS) was used as positive control. LPS significantly increased PGE2 levels in the incubation medium of monocyte cultures after 24 h. Alumina had no effect on PGE2 production, whereas metals induced a concentration-dependent increase in PGE2 release, that was statistically significant at the dose of 0.1 mg/ml. In lymphocytes, LPS elicited a weak but significant increase in PGE2 release, whereas both alumina and metals did not modify PGE2 amounts at any of the concentrations tested. The gene expression of a number of pro- and anti-inflammatory cytokines was assessed in monocytes and lymphocytes exposed to LPS, 0.1 mg/ml alumina or 0.1 mg/ml metals for 24 h. In monocytes, LPS caused a 2-fold increase in interleukin-1beta (IL-1beta) mRNA levels. The exposure of monocytes to metals resulted in a selective increase in IL-1beta mRNA accumulation (+48% compared to control). By contrast, alumina did not modify IL-1beta mRNA levels. None of the test substances elicited any response on purified lymphocyte population. These findings suggest that PGE2 production and IL-1 mRNA expression are a reliable marker to study the pro-inflammatory effects of wear debris in vitro. The lower activity of alumina compared to metals suggests that the former should be preferred in implants for its favorable biological and mechanical behavior.


Subject(s)
Dinoprostone/metabolism , Gene Expression Regulation/drug effects , Interleukin-1/genetics , Metals/pharmacology , Monocytes/drug effects , Aluminum Oxide/pharmacology , Cell Survival/drug effects , Cells, Cultured , Ceramics/pharmacology , Dose-Response Relationship, Drug , Humans , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Monocytes/cytology , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Neurochem Int ; 49(5): 525-32, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16720060

ABSTRACT

The amyloid beta-peptide (AbetaP) is the major protein component of brain senile plaques in Alzheimer's disease. The redox state of methionine-35 residue plays a critical role in peptide neurotoxic actions. We used the fragment 31-35 of AbetaP [AbetaP(31-35)], containing a single methionine-35 residue (Met-35), to investigate the relationship between the oxidative state of Met-35 and neurotoxic and pro-apoptotic actions induced by the peptide; in rat cerebellar granule cells (CGC), we compared the effects of AbetaP(31-35), in which the Met-35 is present in the reduced state, with those of a modified peptide with oxidized Met-35 [AbetaP(31-35)Met-35(OX)](,) as well as an AbetaP-derivative with Met-35 substituted by norleucine [AbetaP(31-35)Nle-35]. AbetaP(31-35) induced a time-dependent decrease in cell viability. AbetaP(31-35)Met-35(OX) was significantly less potent, but still induced a significant decrease in cell viability compared to control. No toxic effects were observed after treatment with AbetaP(31-35)Nle-35. AbetaP(31-35) induced a 2-fold increase in bax mRNA levels after 4h, whereas AbetaP(31-35)Met-35(OX) raised bax mRNA levels by 41% and AbetaP(31-35)Nle-35 had no effect. Finally, AbetaP(31-35) caused a 43% increase in caspase-3 activity after 24h; AbetaP(31-35)Met-35(OX) caused only a 18% increase, and AbetaP(31-35)Nle-35 had no effect. These findings suggest that AbetaP(31-35)-induced neurodegeneration in CGC is mediated by a selective early increase in bax mRNA levels followed by delayed caspase-3 activation; the redox state of the single Met-35 residue is crucial in the occurrence and extent of the above phenomena.


Subject(s)
Amyloid beta-Peptides/pharmacology , Caspases/metabolism , Cerebellum/drug effects , Cytoplasmic Granules/drug effects , Gene Expression Regulation/drug effects , Methionine/metabolism , Peptide Fragments/pharmacology , bcl-2-Associated X Protein/genetics , Amyloid beta-Peptides/metabolism , Animals , Caspase 3 , Cells, Cultured , Cerebellum/pathology , Enzyme Activation , Oxidation-Reduction , Peptide Fragments/metabolism , Rats
4.
J Neurosci Res ; 83(6): 1058-65, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16609959

ABSTRACT

The endogenous cannabinoid system is involved in the regulation of a number of physiologic effects in both the central and peripheral nervous systems. Its role in the control of neuronal cell proliferation has attracted major attention because of its potential implications for new therapeutic strategies. In the present study, we demonstrated that treatment of cultured cerebellar granule cells with the synthetic cannabinoid WIN55,212-2, causes cell-body and nuclear shrinkage, which are hallmarks of neuronal apoptosis, as well as concentration-dependent decrease in cell viability. Staining with the fluorescent nuclear dye, Hoechst 33258, revealed apoptosis in 27.1% and 58.5% of cells exposed to 1 and 10 microM of WIN55,212-2, respectively (P < 0.01 and P < 0.001 vs. control respectively) after 36 hr. After 24 hr of exposure to WIN55,212-2, mRNA levels for the anti-apoptotic gene bcl-xL were reduced to 45.6% of those found in control (P < 0.01). These effects were completely reverted when cells were exposed to the synthetic cannabinoid in the presence of the specific CB1-receptor antagonist, SR141716A (1 microM). Moreover, the pro-apoptotic effect of 10 microM WIN55,212-2 could be reduced by the addition to the incubation medium of a cell-permeant inhibitor of caspase-1 (50 nM). Finally, WIN55,212-2 significantly increased caspase-1 activity after 24 hr. These findings show that the activation of CB1 receptors on cerebellar granule cells induces apoptotic cell death, which is associated with downregulation of the anti-apoptotic gene, bcl-xL, and at least in part, activation of caspase-1.


Subject(s)
Apoptosis/drug effects , Cannabinoids/agonists , Morpholines/pharmacology , Naphthalenes/pharmacology , Neurons/drug effects , Receptor, Cannabinoid, CB1/physiology , bcl-X Protein/metabolism , Analysis of Variance , Animals , Animals, Newborn , Benzoxazines , Blotting, Western/methods , Cell Count/methods , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Interactions , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Rats , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant , Time Factors , bcl-X Protein/genetics
5.
J Neuroimmunol ; 160(1-2): 61-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15710458

ABSTRACT

The effects of interleukin-18 (IL-18), a putative member of the IL-1 family, were investigated on basal and stimulated release of corticotropin-releasing hormone (CRH) and prostanoids from rat hypothalamic explants and glial cells in vitro. We found that IL-18 decreases basal and KCl-stimulated CRH release from the hypothalamus. IL-18 also reduced CRH gene expression after 1- and 3-h incubation. The cytokine did not modify basal PGE2 production by hypothalamic explants but abolished production stimulated by IL-1beta. Similar effects were also observed on cultured glial cells. The present findings show that IL-18 possesses a profile of in vitro neuroendocrine activities opposing to, and even antagonizing, those of IL-1beta.


Subject(s)
Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/immunology , Interleukin-18/physiology , Interleukin-1/antagonists & inhibitors , Animals , Astrocytes/immunology , Astrocytes/metabolism , Cells, Cultured , Corticotropin-Releasing Hormone/genetics , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Dose-Response Relationship, Immunologic , Gene Expression Regulation/immunology , Humans , Hypothalamus/cytology , Hypothalamus/metabolism , Interleukin-1/physiology , Male , Microglia/immunology , Microglia/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Stress, Physiological/immunology , Stress, Physiological/metabolism , Stress, Physiological/physiopathology
6.
Toxicol Lett ; 148(1-2): 91-4, 2004 Mar 14.
Article in English | MEDLINE | ID: mdl-15019092

ABSTRACT

Toxicity of anticancer agents on normal neural cells during chemotherapy of primary or secondary brain tumors is a clinical problem of increasing relevance and concern. In this perspective, here we used primary cultures of rat cortical microglia as an in vitro paradigm of normal glia to investigate the neurotoxicity of anticancer agents. The effects of two compounds frequently used for treatment of brain tumors, methotrexate (MTX) and temozolomide (TMZ), were compared to those of a known microglial activator, bacterial lipopolysaccharide (LPS); cell viability and metabolism was assessed by the MTS assay. We found that LPS, in the low-intermediate range of concentrations, strongly activates microglia cells, but a highly significant decrease in viability was observed from 100 ng/ml onward. TMZ has no effect at concentrations of clinical interest, whereas MTX significantly increases cell metabolism at 30 microM, a phenomenon possibly reflecting MTX neurotoxicity observed in patients.


Subject(s)
Antineoplastic Agents/adverse effects , Dacarbazine/analogs & derivatives , Drug Screening Assays, Antitumor , Microglia/drug effects , Animals , Animals, Newborn , Antineoplastic Agents, Alkylating/adverse effects , Cell Culture Techniques , Cell Survival , Dacarbazine/adverse effects , Humans , Lipopolysaccharides/toxicity , Methotrexate/adverse effects , Nervous System Diseases/chemically induced , Rats , Rats, Wistar , Temozolomide , Toxicity Tests/methods
7.
Glia ; 45(4): 354-63, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966867

ABSTRACT

Changes in intracellular Ca2+ levels are an important signal underlying neuron-glia cross-talk, but little is known about the possible role of voltage-gated Ca2+ channels (VGCCs) in controlling glial cell Ca2+ influx. We investigated the pharmacological and biophysical features of VGCCs in cultured rat cortical astrocytes. In whole-cell patch-clamp experiments, L-channel blockade (5 microM nifedipine) reduced Ba2+ current amplitude by 28% of controls, and further decrease (32%) was produced by N-channel blockade (3 microM omega-conotoxin-GVIA). No significant additional changes were observed after P/Q channel blockade (3 microM omega-conotoxin-MVIIC). Residual current (36% of controls) amounted to roughly the same percentage (34%) that was abolished by R-channel blockade (100 nM SNX-482). Electrophysiological evidence of L-, N-, and R-channels was associated with RT-PCR detection of mRNA transcripts for VGCC subunits alpha1C (L-type), alpha1B (N-type), and alpha1E (R-type). In cell-attached recordings, single-channel properties (L-currents: amplitude, -1.21 +/- 0.02 pA at 10 mV; slope conductance, 22.0 +/- 1.1 pS; mean open time, 5.95 +/- 0.24 ms; N-currents: amplitude, -1.09 +/- 0.02 pA at 10 mV; slope conductance, 18.0 +/- 1.1 pS; mean open time, 1.14 +/- 0.02 ms; R-currents: amplitude, -0.81 +/- 0.01 pA at 20 mV; slope conductance, 10.5 +/- 0.3 pS; mean open time, 0.88 +/- 0.02 ms) resembled those of corresponding VGCCs in neurons. These novel findings indicate that VGCC expression by cortical astrocytes may be more varied than previously thought, suggesting that these channels may indeed play substantial roles in the regulation of astrocyte Ca2+ influx, which influences neuron-glia cross-talk and numerous other calcium-mediated glial-cell functions.


Subject(s)
Astrocytes/physiology , Calcium Channels, L-Type/physiology , Calcium Channels, N-Type/physiology , Calcium Channels, R-Type/physiology , Cerebral Cortex/physiology , Animals , Astrocytes/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/biosynthesis , Calcium Channels, L-Type/genetics , Calcium Channels, N-Type/biosynthesis , Calcium Channels, N-Type/genetics , Calcium Channels, R-Type/biosynthesis , Calcium Channels, R-Type/genetics , Cells, Cultured , Cerebral Cortex/drug effects , Electrophysiology , Rats , Rats, Wistar
8.
Neurosci Lett ; 354(2): 107-10, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14698450

ABSTRACT

Statins were recently shown to possess anti-inflammatory activities, which might be responsible for their favourable effects in cardiovascular or CNS disorders independently from the inhibition of hydroxy-methyl-glutaryl CoA reductase. Here we investigated the effects of the statins lovastatin and mevastatin on prostanoid production in primary cultures of rat cortical microglia and astrocytes. We found that both statins significantly reduce prostaglandin E2 (PGE2) release from microglia, either under basal conditions or after stimulation by interleukin-1beta. Lovastatin also tends to reduce, although not in a significant manner, basal and interleukin-1beta-stimulated PGE2 release from astrocytes. Precursors and intermediates in cholesterol biosynthesis--mevalonic acid and geranyl and farnesyl pyrophosphate--also reduce PGE2 production, and potentiate the inhibitory effects of statins, suggesting that the latter might not depend on the inhibition of hydroxy-methyl-glutaryl CoA reductase.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/analogs & derivatives , Microglia/drug effects , Prostaglandins/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/physiopathology , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Synergism , Encephalitis/drug therapy , Encephalitis/metabolism , Encephalitis/physiopathology , Interleukin-1/pharmacology , Lovastatin/pharmacology , Mevalonic Acid/metabolism , Mevalonic Acid/pharmacology , Microglia/cytology , Microglia/metabolism , Polyisoprenyl Phosphates/metabolism , Polyisoprenyl Phosphates/pharmacology , Rats , Sesquiterpenes
9.
J Psychiatry Neurosci ; 29(6): 459-66, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15644987

ABSTRACT

OBJECTIVE: Corticotropin-releasing factor (CRF), the major adrenocorticotropic hormone (ACTH) secretagogue, acts within the brain to integrate the stress responses of the central nervous, endocrine and immune systems. The involvement of this peptide in the origin and pathophysiology of various endocrine, neurologic, inflammatory and psychiatric diseases, particularly affective disorders, has also been suggested. The antiepileptic drug valproic acid is frequently used as a mood-stabilizing agent in patients with bipolar disorders; however, its mechanism of action for the latter indication is still poorly characterized. We investigated whether valproic acid can directly modulate CRF production by using the incubation of rat hypothalamic explants as an in-vitro model. We then studied the involvement of the gamma-aminobutyric acid (GABA) system as a putative mediator of the effects of valproic acid on CRF production. METHODS: Rat hypothalamic explants were incubated in a 24-well plate (2 hypothalami per well) at 37 degrees C in a humidified atmosphere (5; CO(2) and 95% O(2)) in incubation medium, 700 muL, then were treated with medium alone (control) or test substances, namely, valproic acid, KCI, bicuculline methiodide and muscimol. Released CRF was measured by radioimmunoassay. CRF mRNA was measured by RNase protection analysis. RESULTS: Incubation of the hypothalamic fragments with valproic acid, 100 mumol/L, resulted in a reduction of basal CRF secretion after 3 hours' treatment. The drug was also able to inhibit KCl-stimulated CRF release. Moreover, valproic acid, 100 mumol/L, significantly decreased CRF mRNA levels after 3 hours. A specific GABA(A) receptor antagonist, bicuculline methiodide, completely reversed the inhibition of CRF gene expression and peptide release induced by valproic acid; in this paradigm, the GABA(A)-specific agonist muscimol inhibited both CRF gene expression and peptide release in a concentration-dependent manner. CONCLUSIONS: These results suggest that valproic acid may exert part of its therapeutic effect as a mood-stabilizing drug via the modulation of CRF secretion from the hypothalamus. This action may be mediated in part by the activation of GABAergic neurotransmission.


Subject(s)
Anticonvulsants/pharmacology , Bicuculline/analogs & derivatives , Corticotropin-Releasing Hormone/metabolism , Hippocampus/drug effects , Synaptic Transmission/drug effects , Valproic Acid/pharmacology , gamma-Aminobutyric Acid/physiology , Animals , Bicuculline/pharmacology , Corticotropin-Releasing Hormone/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hippocampus/pathology , Male , Muscimol/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Secretory Rate/drug effects , Tissue Culture Techniques
10.
Eur J Neurosci ; 16(4): 584-92, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12270034

ABSTRACT

Erythropoietin (EPO), a renal cytokine regulating haematopoiesis, is also produced by different cell types within the central nervous system, where it acts via the activation of specific receptors. Current evidence shows that EPO exerts neurotrophic and neuroprotective activities in different in vivo and in vitro models of brain damage. In the present study we investigated the effects of EPO on primary cultures of rat cortical microglia and astrocytes. We found that: (i) EPO exerted a marked stimulatory effect on microglial cell viability, assessed through the MTS assay, whereas astrocytes were almost unaffected; (ii) the cytokine increased microglial cell population size in a concentration-dependent manner; however, as microglia cultures undergo spontaneous apoptosis after separation from astrocytes, the apparent effect on cell proliferation could be attributed to EPO antagonism of normal apoptosis; (iii) subsequent flow cytometry analysis on microglial cells demonstrated both the trophic role of factor(s) released by astrocytes in mixed cultures, and the putative anti-apoptotic action of EPO; (iv) the latter was further confirmed through the assessment of gene expression of anti- and pro-apoptotic factors, which showed that EPO is able to shift the Bcl : Bax ratio towards a net anti-apoptotic effect; (v) EPO did not affect the pro-inflammatory function of microglial cells.


Subject(s)
Apoptosis/drug effects , Erythropoietin/pharmacology , Microglia/drug effects , Animals , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Erythropoietin/physiology , Humans , Microglia/cytology , Microglia/physiology , Rats , Rats, Wistar , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...