Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
JHEP Rep ; 1(3): 145-153, 2019 Sep.
Article in English | MEDLINE | ID: mdl-32039364

ABSTRACT

BACKGROUND & AIMS: Patients with primary biliary cholangitis (PBC) exhibit reduced AE2/SLC4A2 gene expression in the liver and peripheral blood mononuclear cells (PBMCs). AE2 encodes a Cl-/HCO3 - exchanger involved in biliary bicarbonate secretion and intracellular pH regulation. Reduced AE2 expression in PBC may be pathogenic, as Ae2-knockout mice reproduce characteristic PBC features. Herein, we aimed to identify CpG-methylation abnormalities in AE2 promoter regions that might contribute to the reduced gene transcription in PBC livers and PBMCs. METHODS: CpG-cytosine methylation rates were interrogated at 1-base pair resolution in upstream and alternate AE2 promoter regions through pyrosequencing of bisulphite-modified genomic DNA from liver specimens and PBMCs. AE2a and alternative AE2b1 and AE2b2 mRNA levels were measured by real-time PCR. Human lymphoblastoid-T2 cells were treated with 5-aza-2´-deoxycytidine for demethylation assays. RESULTS: AE2 promoters were found to be hypermethylated in PBC livers compared to normal and diseased liver specimens. Receiver operating characteristic (ROC) curve analysis showed that minimal CpG-hypermethylation clusters of 3 AE2a-CpG sites and 4 alternate-AE2b2-CpG sites specifically differentiated PBC from normal and diseased controls, with mean methylation rates inversely correlating with respective transcript levels. Additionally, in PBMCs a minimal cluster of 3 hypermethylated AE2a-CpG sites distinguished PBC from controls, and mean methylation rates correlated negatively with AE2a mRNA levels in these immune cells. Alternate AE2b2/AE2b1 promoters in PBMCs were constitutively hypermethylated, in line with absent alternative mRNA expression in diseased and healthy PBMCs. Demethylation assays treating lymphoblastoid-T2 cells with 5-aza-2´-deoxycytidine triggered AE2b2/AE2b1 expression and upregulated AE2a-promoter expression. CONCLUSIONS: Disease-specific hypermethylation of AE2 promoter regions and subsequent downregulation of AE2-gene expression in the liver and PBMCs of patients with PBC might be critically involved in the pathogenesis of this complex disease. LAY SUMMARY: Primary biliary cholangitis (PBC) is a chronic immune-associated cholestatic liver disease with unclear complex/multifactorial etiopathogenesis affecting mostly middle-aged women. Patients with PBC exhibit reduced expression of the AE2/SLC4A2 gene. Herein, we found that AE2 promoter regions are hypermethylated in the liver and peripheral blood mononuclear cells of patients with PBC. This increased methylation is associated with downregulated AE2-gene expression, which might contribute to the pathogenesis of PBC. Therefore, novel epigenetic targets may improve treatment in patients with PBC who respond poorly to current pharmacological therapies.

2.
Biochim Biophys Acta ; 1832(6): 705-17, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23415811

ABSTRACT

Phosphodiesterase 5 (PDE5) inhibitors have recently been reported to exert beneficial effects against ischemia-reperfusion injury in several organs but their neuroprotective effects in brain stroke models are scarce. The present study was undertaken to assess the effects of sildenafil against cell death caused by intrastriatal injection of malonate, an inhibitor of succinate dehydrogenase; which produces both energy depletion and lesions similar to those seen in cerebral ischemia. Our data demonstrate that sildenafil (1.5mg/kg by mouth (p.o.)), given 30min before malonate (1.5µmol/2µL), significantly decreased the lesion volume caused by this toxin. This protective effect can be probably related to the inhibition of excitotoxic pathways. Thus, malonate induced the activation of the calcium-dependent protease, calpain and the cyclin-dependent kinase 5, cdk5; which resulted in the hyperphosphorylation of tau and the cleavage of the protective transcription factor, myocyte enhancer factor 2, MEF2. All these effects were also significantly reduced by sildenafil pre-treatment, suggesting that sildenafil protects against malonate-induced cell death through the regulation of the calpain/p25/cdk5 signaling pathway. Similar findings were obtained using inhibitors of calpain or cdk5, further supporting our contention. Sildenafil also increased MEF2 phosphorylation and Bcl-2/Bax and Bcl-xL/Bax ratios, effects that might as well contribute to prevent cell death. Finally, sildenafil neuroprotection was extended not only to rat hippocampal slices subjected to oxygen and glucose deprivation when added at the time of reoxygenation, but also, in vivo when administered after malonate injection. Thus, the therapeutic window for sildenafil against malonate-induced hypoxia was set at 3h.


Subject(s)
Calpain/metabolism , Cyclin-Dependent Kinase 5/metabolism , Hypoxia, Brain , Malonates/toxicity , Neuroprotective Agents/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Hypoxia, Brain/chemically induced , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Hypoxia, Brain/prevention & control , Male , Phosphorylation/drug effects , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction/drug effects , Sildenafil Citrate , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , tau Proteins/metabolism
3.
J Neurosci Res ; 90(2): 518-28, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21948520

ABSTRACT

Sildenafil, given shortly before 3,4-methylenedioxymethamphetamine (MDMA), affords protection against 5-hydroxytryptamine (5-HT) depletions caused by this amphetamine derivative by an acute preconditioning-like mechanism. Because acute and delayed preconditionings do not share the same mechanisms, we investigated whether sildenafil would also protect the 5-HT system of the rat if given 24 hr before MDMA. For this, MDMA (3 × 5 mg/kg i.p., every 2 hr) was administered to rats previously treated with sildenafil (8 mg/kg p.o.). One week later, 5-HT content and 5-HT transporter density were measured in the striatum, frontal cortex, and hippocampus of the rats. Our findings indicate that sildenafil afforded significant protection against MDMA-induced 5-HT deficits without altering the acute hyperthermic response to MDMA or its metabolic disposition. Sildenafil promoted ERK1/2 activation an effect that was paralleled by an increase in MnSOD expression that persisted 24 hr later. In addition, superoxide and superoxide-derived oxidants, shown by ethidium fluorescence, increased after the last MDMA injection, an effect that was prevented by sildenafil pretreatment. Similarly, MDMA increased nitrotyrosine concentration in the hippocampus, an effect not shown by sildenafil-pretreated rats. In conclusion, our data demonstrate that sildenafil produces a significant, long-lasting neuroprotective effect against MDMA-induced 5-HT deficits. This effect is apparently mediated by an increased expression of MnSOD and a subsequent reduced susceptibility to the oxidative stress caused by MDMA.


Subject(s)
Brain/drug effects , Brain/pathology , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neuroprotective Agents/administration & dosage , Piperazines/administration & dosage , Serotonin/deficiency , Sulfones/administration & dosage , Animals , Brain/metabolism , Male , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , Purines/administration & dosage , Rats , Rats, Wistar , Serotonin/metabolism , Sildenafil Citrate , Time Factors
4.
J Neurosci ; 31(44): 15826-37, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22049426

ABSTRACT

Mutations in Cu,Zn superoxide dismutase (SOD1) are associated with familial amyotrophic lateral sclerosis (ALS). Mutant SOD1 causes a complex array of pathological events, through toxic gain of function mechanisms, leading to selective motor neuron degeneration. Mitochondrial dysfunction is among the well established toxic effects of mutant SOD1, but its mechanisms are just starting to be elucidated. A portion of mutant SOD1 is localized in mitochondria, where it accumulates mostly on the outer membrane and inside the intermembrane space (IMS). Evidence in cultured cells suggests that mutant SOD1 in the IMS causes mitochondrial dysfunction and compromises cell viability. Therefore, to test its pathogenic role in vivo we generated transgenic mice expressing G93A mutant or wild-type (WT) human SOD1 targeted selectively to the mitochondrial IMS (mito-SOD1). We show that mito-SOD1 is correctly localized in the IMS, where it oligomerizes and acquires enzymatic activity. Mito-G93ASOD1 mice, but not mito-WTSOD1 mice, develop a progressive disease characterized by body weight loss, muscle weakness, brain atrophy, and motor impairment, which is more severe in females. These symptoms are associated with reduced spinal motor neuron counts and impaired mitochondrial bioenergetics, characterized by decreased cytochrome oxidase activity and defective calcium handling. However, there is no evidence of muscle denervation, a cardinal pathological feature of ALS. Together, our findings indicate that mutant SOD1 in the mitochondrial IMS causes mitochondrial dysfunction and neurodegeneration, but per se it is not sufficient to cause a full-fledged ALS phenotype, which requires the participation of mutant SOD1 localized in other cellular compartments.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/ultrastructure , Mitochondria , Mutation/genetics , Spinal Cord/ultrastructure , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Analysis of Variance , Animals , Body Weight/genetics , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Energy Metabolism/genetics , Heart , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Muscle, Skeletal/pathology , Myocardium/pathology , Nerve Tissue Proteins/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
5.
J Neurochem ; 114(3): 843-52, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20477949

ABSTRACT

The aim of the present study was to investigate whether late pre-conditioning using 3-nitropropionic acid (3NP) prevents the 5-hydroxytryptamine (5-HT) deficits caused by the amphetamine derivative 3,4-methylenedioxymethamphetamine (MDMA) in the rat. For this purpose we administered 3NP 24 h before MDMA (3 x 5 mg/kg i.p., every 2 h) and rats were killed 7 days later. Pre-treatment of 3NP afforded complete protection against MDMA-induced 5-HT deficits independent of any effect on MDMA-induced hyperthermia or 5-HT transporter activity. To identify the transductional mechanisms responsible for the neuroprotective effect of 3NP, we first examined the involvement of nitric oxide (NO) by using selective inhibitors of all three nitric oxide synthase isoforms. Inhibition of endothelial and neuronal nitric oxide synthase, but not inducible nitric oxide synthase, reversed 3NP-induced pre-conditioning. The NO donor S-Nitroso-N-acetylpenicilamine mimicked 3NP effects further suggesting the involvement of NO in mediating 3NP protection. To investigate the involvement of NOS/soluble guanylate cyclase (sGC)/protein kinase G/mitochondrial ATP-sensitive potassium channels (mitoK(ATP)) signaling pathway we examined the effect of 5-hydroxydecanoate (5-HD), a selective mitoK(ATP) blocker, and 1H-(1,2,4)oxadiazolo[4,3-a]quinoxaline-1-one, a potent inhibitor of sGC, on 3NP-induced tolerance. 5-hydroxydecanoate, but not 1H-(1,2,4)oxadiazolo[4,3-a]quinoxaline-1-one, suppressed 3NP-mediated protection suggesting that mitoK(ATP) opening, but not NO-mediated activation of sGC, participates in the mechanism underlying tolerance to MDMA. Our data also showed that the protective effect of 3NP was abolished by cycloheximide, supporting the involvement of de novo protein synthesis. In conclusion, 3NP-induced delayed tolerance against 5-HT deficits caused by MDMA occurs via NO production.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neuroprotective Agents/pharmacology , Nitro Compounds/pharmacology , Propionates/pharmacology , Serotonin/deficiency , Animals , Male , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Neuroprotective Agents/administration & dosage , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitro Compounds/administration & dosage , Propionates/administration & dosage , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Wistar , Serotonin/toxicity , Serotonin Agents/administration & dosage , Serotonin Agents/pharmacology
6.
Br J Pharmacol ; 160(2): 233-45, 2010 May.
Article in English | MEDLINE | ID: mdl-20423338

ABSTRACT

BACKGROUND AND PURPOSE: 3,4-methylenedioxymethamphetamine (MDMA) causes a persistent loss of dopaminergic cell bodies in the substantia nigra of mice. Current evidence indicates that such neurotoxicity is due to oxidative stress but the source of free radicals remains unknown. Inhibition of mitochondrial electron transport chain complexes by MDMA was assessed as a possible source. EXPERIMENTAL APPROACH: Activities of mitochondrial complexes after MDMA were evaluated spectrophotometrically. In situ visualization of superoxide production in the striatum was assessed by ethidium fluorescence and striatal dopamine levels were determined by HPLC as an index of dopaminergic toxicity. KEY RESULTS: 3,4-methylenedioxymethamphetamine decreased mitochondrial complex I activity in the striatum of mice, an effect accompanied by an increased production of superoxide radicals and the inhibition of endogenous aconitase. alpha-Lipoic acid prevented superoxide generation and long-term toxicity independent of any effect on complex I inhibition. These effects of alpha-lipoic acid were also associated with a significant increase of striatal glutathione levels. The relevance of glutathione was supported by reducing striatal glutathione content with L-buthionine-(S,R)-sulfoximine, which exacerbated MDMA-induced dopamine deficits, effects suppressed by alpha-lipoic acid. The nitric oxide synthase inhibitor, N(G)-nitro-L-arginine, partially prevented MDMA-induced dopamine depletions, an effect reversed by L-arginine but not D-arginine. Finally, a direct relationship between mitochondrial complex I inhibition and long-term dopamine depletions was found in animals treated with MDMA in combination with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. CONCLUSIONS AND IMPLICATIONS: Inhibition of mitochondrial complex I following MDMA could be the source of free radicals responsible for oxidative stress and the consequent neurotoxicity of this drug in mice.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Hallucinogens/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurotoxicity Syndromes/etiology , Animals , Antioxidants/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Free Radicals/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Thioctic Acid/pharmacology
7.
Neurobiol Dis ; 38(2): 237-45, 2010 May.
Article in English | MEDLINE | ID: mdl-20109548

ABSTRACT

In this study we tested whether phosphodiesterase 5 (PDE5) inhibitors, sildenafil and vardenafil, would afford protection against 3-nitropropionic acid (3NP), which produces striatal lesions that closely mimic some of the neuropathological features of Huntington's Disease (HD). The neurotoxin was given over 5 days by constant systemic infusion using osmotic minipumps. Animals treated with PDE5 inhibitors (sildenafil or vardenafil) showed improved neurologic scores, reduced the loss of striatal DARPP-32 protein levels and lesion volumes, and decreased calpain activation produced by 3NP. This protective effect was independent of changes in 3NP-induced succinate dehydrogenase inhibition. Furthermore, striatal p-CREB levels along with the expression of BDNF were significantly increased in sildenafil-treated rats. In summary, PDE5 inhibitors protected against 3NP-induced striatal degeneration by reducing calpain activation and by promoting survival pathways. These data encourage further evaluation of PDE5 inhibitors in transgenic mouse models of HD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Calpain/metabolism , Corpus Striatum/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Neurotoxicity Syndromes/drug therapy , Nitro Compounds/pharmacology , Piperazines/pharmacology , Propionates/pharmacology , Sulfones/pharmacology , Analysis of Variance , Animals , Blotting, Western , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Male , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Neurotoxins/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Purines/pharmacology , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Succinate Dehydrogenase/metabolism
8.
Hum Mol Genet ; 18(23): 4552-64, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19779023

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by motor neuron degeneration. Mutations in Cu,Zn-superoxide dismutase (SOD1) are responsible for 20% of familial ALS cases via a toxic gain of function. In mutant SOD1 transgenic mice, mitochondria of spinal motor neurons develop abnormal morphology, bioenergetic defects and degeneration, which are presumably implicated in disease pathogenesis. SOD1 is mostly a cytosolic protein, but a substantial portion is associated with organelles, including mitochondria, where it localizes predominantly in the intermembrane space (IMS). However, whether mitochondrial mutant SOD1 contributes to disease pathogenesis remains to be elucidated. We have generated NSC34 motor neuronal cell lines expressing wild-type or mutant SOD1 containing a cleavable IMS targeting signal to directly investigate the pathogenic role of mutant SOD1 in mitochondria. We show that mitochondrially-targeted SOD1 localizes to the IMS, where it is enzymatically active. We prove that mutant IMS-targeted SOD1 causes neuronal toxicity under metabolic and oxidative stress conditions. Furthermore, we demonstrate for the first time neurite mitochondrial fragmentation and impaired mitochondrial dynamics in motor neurons expressing IMS mutant SOD1. These defects are associated with impaired maintenance of neuritic processes. Our findings demonstrate that mutant SOD1 localized in the IMS is sufficient to determine mitochondrial abnormalities and neuronal toxicity, and contributes to ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Mitochondria/enzymology , Motor Neurons/enzymology , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase/toxicity , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondrial Membranes/enzymology , Motor Neurons/pathology , Oxidative Stress , Protein Transport , Superoxide Dismutase-1
9.
J Neurochem ; 108(3): 755-66, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187094

ABSTRACT

Phosphodiesterase 5 (PDE5) inhibitors are often used in combination with club drugs such as 3,4-methylenedioxymethamphetamine (MDMA or ecstasy). We investigated the consequences of such combination in the serotonergic system of the rat. Oral administration of sildenafil citrate (1.5 or 8 mg/kg) increased brain cGMP levels and protected in a dose-dependent manner against 5-hydroxytryptamine depletions caused by MDMA (3 x 5 mg/kg, i.p., every 2 h) in the striatum, frontal cortex and hippocampus without altering the acute hyperthermic response to MDMA. Intrastriatal administration of the protein kinase G (PKG) inhibitor, KT5823 [(9S, 10R, 12R)-2,3,9,10,11,12-Hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid, methyl ester)], suppressed sildenafil-mediated protection. By contrast, the cell permeable cGMP analogue, 8-bromoguanosine cyclic 3',5'-monophosphate, mimicked sildenafil effects further suggesting the involvement of the PKG pathway in mediating sildenafil protection. Because mitochondrial ATP-sensitive K(+) channels are a target for PKG, we next administered the specific mitochondrial ATP-sensitive K(+) channel blocker, 5-hydroxydecanoic acid, 30 min before sildenafil. 5-hydroxydecanoic acid completely reversed the protection afforded by sildenafil, thereby implicating the involvement of mitochondrial ATP-sensitive K(+) channels. Sildenafil also increased Akt phosphorylation, and so the possible involvement of the Akt/endothelial nitric oxide synthase (eNOS)/sGC signalling pathway was analysed. Neither the phosphatidylinositol 3-kinase inhibitor, wortmannin, nor the selective eNOS inhibitor, L-N5-(1-iminoethyl)-L-ornithine dihydrochloride, reversed the protection afforded by sildenafil, suggesting that Akt/eNOS/sGC cascade does not participate in the protective mechanisms. Our data also show that the protective effect of sildenafil can be extended to vardenafil, another PDE5 inhibitor. In conclusion, sildenafil protects against MDMA-induced long-term reduction of indoles by a mechanism involving increased production of cGMP and subsequent activation of PKG and mitochondrial ATP-sensitive K(+) channel opening.


Subject(s)
Hallucinogens/antagonists & inhibitors , Hallucinogens/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Serotonin/deficiency , Sulfones/pharmacology , Animals , Blotting, Western , Body Temperature/drug effects , Brain Chemistry/drug effects , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Hydroxyindoleacetic Acid/metabolism , Imidazoles/pharmacology , KATP Channels/drug effects , Male , Microinjections , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/drug effects , Purines/pharmacology , Rats , Rats, Wistar , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction/drug effects , Sildenafil Citrate , Triazines/pharmacology , Vardenafil Dihydrochloride
10.
Neuropharmacology ; 54(5): 885-900, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18329670

ABSTRACT

The mechanisms underlying 3,4-methylenedioxymethamphetamine (MDMA)-induced serotonergic (5-HT) toxicity remain unclear. It has been suggested that MDMA depletes 5-HT by increasing brain tyrosine levels, which via non-enzymatic hydroxylation leads to DA-derived free radical formation. Because this hypothesis assumes the pre-existence of hydroxyl radicals, we hypothesized that MDMA metabolism into pro-oxidant compounds is the limiting step in this process. Acute hyperthermia, plasma tyrosine levels and concentrations of MDMA and its main metabolites were higher after a toxic (15 mg/kg i.p.) vs. a non-toxic dose of MDMA (7.5mg/kg i.p.). The administration of a non-toxic dose of MDMA in combination with l-tyrosine (0.2 mmol/kg i.p.) produced a similar increase in serum tyrosine levels to those found after a toxic dose of MDMA; however, brain 5-HT content remained unchanged. The non-toxic dose of MDMA combined with a high dose of tyrosine (0.5 mmol/kg i.p.), caused long-term 5-HT depletions in rats treated at 21.5 degrees C but not in those treated at 15 degrees C, conditions known to decrease MDMA metabolism. Furthermore, striatal perfusion of MDMA (100 microM for 5h) combined with tyrosine (0.5 mmol/kg i.p.) in hyperthermic rats did not cause 5-HT depletions. By contrast, rats treated with the non-toxic dose of MDMA under heating conditions or combined with entacapone or acivicin, which interfere with MDMA metabolism or increase brain MDMA metabolite availability respectively, showed significant reductions of brain 5-HT content. Altogether, these data indicate that although tyrosine may contribute to MDMA-induced toxicity, MDMA metabolism appears to be the limiting step.


Subject(s)
Brain/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurotoxicity Syndromes/metabolism , Serotonin/metabolism , Tyrosine/metabolism , Analysis of Variance , Animals , Antimetabolites/pharmacology , Area Under Curve , Body Temperature/drug effects , Brain/metabolism , Brain/pathology , Catechols/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Hydroxyindoleacetic Acid/metabolism , Isoxazoles/pharmacology , Male , Neurotoxicity Syndromes/etiology , Nitriles/pharmacology , Protein Binding/drug effects , Rats , Rats, Wistar , Time Factors , Tyrosine/pharmacology
11.
J Clin Invest ; 118(2): 695-709, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18188457

ABSTRACT

Primary biliary cirrhosis (PBC) is a cholestatic disease associated with autoimmune phenomena and alterations in both biliary bicarbonate excretion and expression of the bicarbonate carrier AE2. The bile acid ursodeoxycholic acid (UCDA) is currently used in treatment of cholestatic liver diseases and is the treatment of choice in PBC; however, a subset of PBC patients respond poorly to UDCA monotherapy. In these patients, a combination of UDCA and glucocorticoid therapy appears to be beneficial. To address the mechanism of this benefit, we analyzed the effects of UDCA and dexamethasone on AE2 gene expression in human liver cells from hepatocyte and cholangiocyte lineages. The combination of UDCA and dexamethasone, but not UDCA or dexamethasone alone, increased the expression of liver-enriched alternative mRNA isoforms AE2b1 and AE2b2 and enhanced AE2 activity. Similar effects were obtained after replacing UDCA with UDCA conjugates. In in vitro and in vivo reporter assays, we found that a UDCA/dexamethasone combination upregulated human AE2 alternate overlapping promoter sequences from which AE2b1 and AE2b2 are expressed. In chromatin immunoprecipitation assays, we demonstrated that combination UCDA/dexamethasone treatment induced p300-related interactions between HNF1 and glucocorticoid receptor on the AE2 alternate promoter. Our data provide a potential molecular explanation for the beneficial effects of the combination of UDCA and glucocorticoids in PBC patients with inadequate response to UDCA monotherapy.


Subject(s)
Anion Transport Proteins/genetics , Antiporters/genetics , Cholagogues and Choleretics/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hepatocytes/drug effects , Transcription, Genetic/drug effects , Ursodeoxycholic Acid/pharmacology , Animals , Anion Transport Proteins/analysis , Anion Transport Proteins/metabolism , Antiporters/analysis , Antiporters/metabolism , Cell Line , Cholagogues and Choleretics/therapeutic use , Chromatin Immunoprecipitation , Dexamethasone/therapeutic use , Drug Therapy, Combination , Gene Expression Regulation , Glucocorticoids/therapeutic use , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocytes/metabolism , Humans , Liver Cirrhosis, Biliary/drug therapy , Mice , Mutation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , SLC4A Proteins , Up-Regulation , Ursodeoxycholic Acid/therapeutic use , p300-CBP Transcription Factors/metabolism
12.
Eur J Pharmacol ; 562(3): 198-207, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17320075

ABSTRACT

Amiloride and several of its congeners known to block the Na(+)/Ca(2+) and/or Na(+)/H(+) antiporters potentiate methamphetamine-induced neurotoxicity without altering methamphetamine-induced hyperthermia. We now examine whether amiloride also exacerbates 3,4-methylenedioxymethamphetamine (MDMA)-induced long-term serotonin (5-HT) loss in rats. Amiloride (2.5 mg/kg, every 2 h x 3, i.p.) given at ambient temperature 30 min before MDMA (5 mg/kg, every 2 h x 3, i.p.), markedly exacerbated long-term 5-HT loss. However, in contrast to methamphetamine, amiloride also potentiated MDMA-induced hyperthermia. Fluoxetine (10 mg/kg i.p.) completely protected against 5-HT depletion caused by the MDMA/amiloride combination without significantly altering the hyperthermic response. By contrast, the calcium channel antagonists flunarizine or diltiazem did not afford any protection. Findings with MDMA and amiloride were extended to the highly selective Na(+)/H(+) exchange inhibitor dimethylamiloride, suggesting that the potentiating effects of amiloride are probably mediated by the blockade of Na(+)/H(+) exchange. When the MDMA/amiloride combination was administered at 15 degrees C hyperthermia did not develop and brain 5-HT concentrations remained unchanged 7 days later. Intrastriatal perfusion of MDMA (100 microM for 8 h) in combination with systemic amiloride caused a small depletion of striatal 5-HT content in animals made hyperthermic but not in the striatum of normothermic rats. These data suggest that enhancement of MDMA-induced 5-HT loss caused by amiloride or dimethylamiloride depends on their ability to enhance MDMA-induced hyperthermia. We hypothesise that blockade of Na(+)/H(+) exchange could synergize with hyperthermia to render 5-HT terminals more vulnerable to the toxic effects of MDMA.


Subject(s)
Amiloride/pharmacology , Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Agents/pharmacology , Serotonin/metabolism , Sodium Channel Blockers/pharmacology , Amiloride/analogs & derivatives , Animals , Body Temperature/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drug Synergism , Fever/chemically induced , Male , Rats , Rats, Wistar , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/drug effects , Sodium-Hydrogen Exchangers/physiology
13.
Muscle Nerve ; 33(5): 598-608, 2006 May.
Article in English | MEDLINE | ID: mdl-16372325

ABSTRACT

The causes of motor neuron death in amyotrophic lateral sclerosis (ALS) are still unknown. Several lines of evidence suggest that mitochondrial dysfunction may be involved in the pathogenesis of ALS. Biochemical and morphological mitochondrial abnormalities have been demonstrated in postmortem spinal cords of ALS patients. Furthermore, in transgenic mice expressing mutant Cu,Zn-superoxide dismutase (SOD1), the antioxidant enzyme associated with familial ALS (FALS), mitochondrial abnormalities precede the disease onset, suggesting that mitochondrial dysfunction is causally involved in the pathogenesis of SOD1-FALS. Despite this evidence, it is not yet fully understood how mutant SOD1 damages mitochondria. Recent work has demonstrated that a portion of mutant SOD1 is localized in mitochondria, both in transgenic mice and in FALS patients, where it forms proteinaceous aggregates. These findings have opened new avenues of investigation addressing the hypothesis that mutant SOD1 may directly damage mitochondria. Major future challenges will be to better understand the mechanisms and the consequences of mitochondrial dysfunction in ALS. If mitochondrial dysfunction is convincingly involved in ALS pathogenesis, either as a primary cause or as contributing factor, it is likely to become a novel target for therapeutic intervention.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/pathology , Mitochondrial Diseases/complications , Amyotrophic Lateral Sclerosis/genetics , Animals , Apoptosis/physiology , Calcium/metabolism , Humans , Mitochondrial Diseases/genetics , Models, Biological , Mutation , Neurons/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
14.
J Med Chem ; 45(19): 4128-39, 2002 Sep 12.
Article in English | MEDLINE | ID: mdl-12213056

ABSTRACT

It has been proposed that 5-HT(1A) receptor antagonists augment the antidepressant efficacy of selective serotonin (5-HT) reuptake inhibitors. In a search toward new and efficient antidepressants, 1-(aryl)-3-[4-arylpiperazin-1-yl]-1-propane molecular hybrids were designed, synthesized, and evaluated for 5-HT reuptake inhibition and 5-HT(1A) receptor affinity. The design was based in coupling structural moieties related to inhibition of serotonin reuptake, such as benzo[b]thiophene derivatives to arylpiperazines, typical 5-HT(1A) receptor ligands. In binding studies, several compounds showed affinity at the 5-HT transporter and at 5-HT(1A) receptors. Molecular modeling studies predicted the pharmacophore elements required for high affinity binding and the features that enable to discriminate between agonist, partial agonist, or antagonist action at 5-HT(1A) receptors and 5-HT transporter inhibition. Solvent interactions in desolvation prior to the binding step along with enthalpy and enthropy compensations might be responsible to explain agonist, partial agonist, and antagonist character. Hydrogen-bonding capability seems to be important to break hydrogen interhelical hydrogen bonds or alternatively to form other bonds upon ligand binding. Partial agonists and antagonists are unable to do this as the full agonist, which interacts closely by long-range forces or directly. The compounds showing the higher affinity at both the 5-HT transporter (K(i) < 50 nM) and the 5-HT(1A) receptors (K(i) < 20 nM) were further explored for their ability to stimulate [(35)S]GTPgammaS binding or to antagonize 8-hydroxy-2-di-n-propylamino-tetralin (8-OH-DPAT)-stimulated [(35)]GTPgammaS binding to rat hippocampal membranes, an index of agonist/antagonist action at 5-HT(1A) receptors, respectively. Compound 8g exhibited agonist activity (EC(50) = 30 nM) in this assay, whereas compounds 7g and 8h,i behaved as weak partial agonists and 7h-j and 8j,l antagonized the R(+)-8-OH-DPAT-stimulated GTPgammaS binding. Functional characterization was performed by measuring the antagonism to 8-OH-DPAT-induced hypothermia in mice.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Membrane Transport Proteins , Nerve Tissue Proteins , Piperazines/chemical synthesis , Propane/analogs & derivatives , Propane/chemical synthesis , Receptors, Serotonin/drug effects , Selective Serotonin Reuptake Inhibitors/chemical synthesis , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Carrier Proteins/chemistry , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypothermia/chemically induced , In Vitro Techniques , Male , Membrane Glycoproteins/chemistry , Mice , Models, Molecular , Piperazines/chemistry , Piperazines/pharmacology , Propane/chemistry , Propane/pharmacology , Radioligand Assay , Rats , Receptors, Serotonin/chemistry , Receptors, Serotonin, 5-HT1 , Serotonin/metabolism , Serotonin Antagonists/chemical synthesis , Serotonin Antagonists/chemistry , Serotonin Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins , Serotonin Receptor Agonists/chemical synthesis , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...