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










Publication year range
1.
Alcohol Clin Exp Res ; 41(3): 531-540, 2017 03.
Article in English | MEDLINE | ID: mdl-28160299

ABSTRACT

BACKGROUND: In our companion article, we examined the role of MyD88-dependent signaling in ethanol (EtOH) consumption in mice lacking key components of this inflammatory pathway and observed differential effects on drinking. Here, we studied the role of these same signaling components in the acute sedative, intoxicating, and physiological effects of EtOH. Toll-like receptor 4 (TLR4) has been reported to strongly reduce the duration of EtOH-induced sedation, although most studies do not support its direct involvement in EtOH consumption. We examined TLR4 and other MyD88 pathway molecules to determine signaling specificity in acute EtOH-related behaviors. We also studied other GABAergic sedatives to gauge the EtOH specificity and potential role for GABA in EtOH's sedative and intoxicating effects in the mutant mice. METHODS: Loss of righting reflex (LORR) and recovery from motor incoordination were studied following acute injection of EtOH or other sedative drugs in male and female control C57BL/6J mice versus mice lacking CD14, TLR2, TLR4 (C57BL/10ScN), or MyD88. We also examined EtOH-induced hypothermia and blood EtOH clearance in these mice. RESULTS: Male and female mice lacking TLR4 or MyD88 showed reduced duration of EtOH-induced LORR and faster recovery from EtOH-induced motor incoordination in the rotarod test. MyD88 knockout mice had slightly faster recovery from EtOH-induced hypothermia compared to control mice. None of the mutants differed from control mice in the rate of blood EtOH clearance. All of the mutants showed similar decreases in the duration of gaboxadol-induced LORR, but only mice lacking TLR4 were less sensitive to the sedative effects of pentobarbital. Faster recovery from diazepam-induced motor impairment was observed in CD14, TLR4, and MyD88 null mice of both sexes. CONCLUSIONS: TLR4 and MyD88 were key mediators of the sedative and intoxicating effects of EtOH and GABAergic sedatives, indicating a strong influence of TLR4-MyD88 signaling on GABAergic function. Despite the involvement of TLR4 in EtOH's acute behaviors, it did not regulate EtOH consumption in any drinking model as shown in our companion article. Collectively, our studies demonstrate differential effects of TLR-MyD88 components in the acute versus chronic actions of EtOH.


Subject(s)
Ethanol/administration & dosage , Lipopolysaccharide Receptors/deficiency , Myeloid Differentiation Factor 88/deficiency , Reflex, Righting/drug effects , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Animals , Female , GABA Modulators/administration & dosage , Hypnotics and Sedatives/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reflex, Righting/physiology , Rotarod Performance Test/methods
2.
eNeuro ; 3(5)2016.
Article in English | MEDLINE | ID: mdl-27822501

ABSTRACT

Proinflammatory pathways in neuronal and non-neuronal cells are implicated in the acute and chronic effects of alcohol exposure in animal models and humans. The nuclear factor-κB (NF-κB) family of DNA transcription factors plays important roles in inflammatory diseases. The kinase IKKß mediates the phosphorylation and subsequent proteasomal degradation of cytosolic protein inhibitors of NF-κB, leading to activation of NF-κB. The role of IKKß as a potential regulator of excessive alcohol drinking had not previously been investigated. Based on previous findings that the overactivation of innate immune/inflammatory signaling promotes ethanol consumption, we hypothesized that inhibiting IKKß would limit/decrease drinking by preventing the activation of NF-κB. We studied the systemic effects of two pharmacological inhibitors of IKKß, TPCA-1 and sulfasalazine, on ethanol intake using continuous- and limited-access, two-bottle choice drinking tests in C57BL/6J mice. In both tests, TPCA-1 and sulfasalazine reduced ethanol intake and preference without changing total fluid intake or sweet taste preference. A virus expressing Cre recombinase was injected into the nucleus accumbens and central amygdala to selectively knock down IKKß in mice genetically engineered with a conditional Ikkb deletion (IkkbF/F ). Although IKKß was inhibited to some extent in astrocytes and microglia, neurons were a primary cellular target. Deletion of IKKß in either brain region reduced ethanol intake and preference in the continuous access two-bottle choice test without altering the preference for sucrose. Pharmacological and genetic inhibition of IKKß decreased voluntary ethanol consumption, providing initial support for IKKß as a potential therapeutic target for alcohol abuse.


Subject(s)
Alcohol Drinking/metabolism , Amygdala/metabolism , I-kappa B Kinase/antagonists & inhibitors , Nucleus Accumbens/metabolism , Alcohol Deterrents/pharmacology , Alcohol Drinking/pathology , Alcohol Drinking/therapy , Amygdala/drug effects , Amygdala/pathology , Animals , Central Nervous System Depressants/administration & dosage , Disease Models, Animal , Ethanol/administration & dosage , Food Preferences/drug effects , Food Preferences/physiology , Gene Knockdown Techniques , I-kappa B Kinase/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , RNA, Messenger/metabolism , Saccharin
3.
Alcohol Clin Exp Res ; 40(3): 563-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857541

ABSTRACT

BACKGROUND: In the accompanying article, we showed that activation of peroxisome proliferator-activated receptor alpha (PPARα) signaling by fenofibrate and tesaglitazar decreases ethanol (EtOH) consumption in mice. In this study, we determined the role of these PPAR agonists in EtOH-related behaviors and other actions that may be important in regulating EtOH consumption. METHODS: The effects of fenofibrate (150 mg/kg) and tesaglitazar (1.5 mg/kg) were examined on the following responses in male and female C57BL/6J (B6) and B6 × 129S4 mice: preference for saccharin, EtOH-induced conditioned place preference (CPP), conditioned taste aversion (CTA), loss of righting reflex, and withdrawal, acoustic startle reflex, response to novelty, and EtOH clearance. Because the B6 inbred strain usually displays weak EtOH-induced CPP and weak EtOH-induced acute withdrawal, B6 × 129S4 mice were also studied. RESULTS: Fenofibrate and tesaglitazar decreased the novelty response and increased acute EtOH withdrawal severity, and fenofibrate increased EtOH-induced CTA. Two important factors for EtOH consumption (saccharin preference and EtOH-induced CPP) were not altered by fenofibrate or tesaglitazar. EtOH clearance was increased by both fenofibrate and tesaglitazar. Response to novelty, acute withdrawal, and EtOH clearance show sex differences and could contribute to the reduced EtOH consumption following fenofibrate administration. CONCLUSIONS: These studies indicate the complexity of EtOH-dependent and EtOH-independent behaviors that are altered by PPAR agonists and provide evidence for novel behavioral actions of these drugs that may contribute to PPAR-mediated effects on alcohol drinking.


Subject(s)
Alcohol Drinking/drug therapy , Alkanesulfonates/pharmacology , Fenofibrate/pharmacology , PPAR alpha/agonists , PPAR alpha/physiology , Phenylpropionates/pharmacology , Alcohol Drinking/psychology , Alkanesulfonates/therapeutic use , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Female , Fenofibrate/therapeutic use , Locomotion/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Phenylpropionates/therapeutic use , Reflex, Righting/drug effects , Reflex, Righting/physiology , Taste/drug effects , Taste/physiology
4.
Alcohol Clin Exp Res ; 40(3): 553-62, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857685

ABSTRACT

BACKGROUND: Several peroxisome proliferator-activated receptor (PPAR) agonists reduce voluntary alcohol consumption in rodent models, and evidence suggests that PPARα and γ subunits play an important role in this effect. To define the subunit dependence of this action, we tested selective PPARα and α/γ agonists and antagonists in addition to null mutant mice lacking PPARα. METHODS: The effects of fenofibrate (PPARα agonist) and tesaglitazar (PPARα/γ agonist) on continuous and intermittent 2-bottle choice drinking tests were examined in male and female wild-type mice and in male mice lacking PPARα. We compared the ability of MK886 (PPARα antagonist) and GW9662 (PPARγ antagonist) to inhibit the effects of fenofibrate and tesaglitazar in wild-type mice. The estrogen receptor antagonist, tamoxifen, can inhibit PPARγ-dependent transcription and was also studied in male and female mice. RESULTS: Fenofibrate and tesaglitazar reduced ethanol (EtOH) consumption and preference in wild-type mice, but these effects were not observed in mice lacking PPARα. MK886 inhibited the action of fenofibrate, but not tesaglitazer, while GW9662 did not inhibit either agonist. The PPAR agonists were more effective in male mice compared to females, and drinking in the continuous 2-bottle choice test was more sensitive to fenofibrate and tesaglitazar compared to drinking in the intermittent access test. Tamoxifen also reduced EtOH consumption in male mice and this action was inhibited by GW9662, but not MK886, suggesting that it acts by activation of PPARγ. CONCLUSIONS: Our study using selective PPAR agonists, antagonists, and null mutant mice indicates a key role for PPARα in mediating reduced EtOH consumption by fenofibrate and tesaglitazar.


Subject(s)
Alcohol Drinking/drug therapy , PPAR alpha/agonists , PPAR alpha/physiology , PPAR gamma/agonists , PPAR gamma/physiology , Protein Subunits/agonists , Alkanesulfonates/pharmacology , Alkanesulfonates/therapeutic use , Anilides/pharmacology , Animals , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Female , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Indoles/pharmacology , Male , Mice , Mice, 129 Strain , Mice, Knockout , PPAR alpha/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , Phenylpropionates/pharmacology , Phenylpropionates/therapeutic use , Protein Subunits/physiology
5.
Neuropharmacology ; 95: 309-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25839897

ABSTRACT

Gene expression studies identified the interleukin-1 receptor type I (IL-1R1) as part of a pathway associated with a genetic predisposition to high alcohol consumption, and lack of the endogenous IL-1 receptor antagonist (IL-1ra) strongly reduced ethanol intake in mice. Here, we compared ethanol-mediated behaviors in mice lacking Il1rn or Il1r1. Deletion of Il1rn (the gene encoding IL-1ra) increases sensitivity to the sedative/hypnotic effects of ethanol and flurazepam and reduces severity of acute ethanol withdrawal. Conversely, deletion of Il1r1 (the gene encoding the IL-1 receptor type I, IL-1R1) reduces sensitivity to the sedative effects of ethanol and flurazepam and increases the severity of acute ethanol withdrawal. The sedative effects of ketamine and pentobarbital were not altered in the knockout (KO) strains. Ethanol intake and preference were not changed in mice lacking Il1r1 in three different tests of ethanol consumption. Recovery from ethanol-induced motor incoordination was only altered in female mice lacking Il1r1. Mice lacking Il1rn (but not Il1r1) showed increased ethanol clearance and decreased ethanol-induced conditioned taste aversion. The increased ethanol- and flurazepam-induced sedation in Il1rn KO mice was decreased by administration of IL-1ra (Kineret), and pre-treatment with Kineret also restored the severity of acute ethanol withdrawal. Ethanol-induced sedation and withdrawal severity were changed in opposite directions in the null mutants, indicating that these responses are likely regulated by IL-1R1 signaling, whereas ethanol intake and preference do not appear to be solely regulated by this pathway.


Subject(s)
Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Ethanol/pharmacology , Hypnotics and Sedatives/pharmacology , Interleukin 1 Receptor Antagonist Protein/metabolism , Receptors, Interleukin-1 Type I/metabolism , Alcohol Drinking/metabolism , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/physiology , Female , Flurazepam/pharmacology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/pharmacology , Ketamine/pharmacology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Pentobarbital/pharmacology , Receptors, Interleukin-1 Type I/genetics , Severity of Illness Index , Substance Withdrawal Syndrome/metabolism , Taste Perception/drug effects , Taste Perception/physiology
6.
J Pharmacol Exp Ther ; 353(1): 181-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25678534

ABSTRACT

Glycine receptors (GlyRs) are broadly expressed in the central nervous system. Ethanol enhances the function of brain GlyRs, and the GlyRα1 subunit is associated with some of the behavioral actions of ethanol, such as loss of righting reflex. The in vivo role of GlyRα2 and α3 subunits in alcohol responses has not been characterized despite high expression levels in the nucleus accumbens and amygdala, areas that are important for the rewarding properties of drugs of abuse. We used an extensive panel of behavioral tests to examine ethanol actions in mice lacking Glra2 (the gene encoding the glycine receptor alpha 2 subunit) or Glra3 (the gene encoding the glycine receptor alpha 3 subunit). Deletion of Glra2 or Glra3 alters specific ethanol-induced behaviors. Glra2 knockout mice demonstrate reduced ethanol intake and preference in the 24-hour two-bottle choice test and increased initial aversive responses to ethanol and lithium chloride. In contrast, Glra3 knockout mice show increased ethanol intake and preference in the 24-hour intermittent access test and increased development of conditioned taste aversion to ethanol. Mutants and wild-type mice consumed similar amounts of ethanol in the limited access drinking in the dark test. Other ethanol effects, such as anxiolysis, motor incoordination, loss of righting reflex, and acoustic startle response, were not altered in the mutants. The behavioral changes in mice lacking GlyRα2 or α3 subunits were distinct from effects previously observed in mice with knock-in mutations in the α1 subunit. We provide evidence that GlyRα2 and α3 subunits may regulate ethanol consumption and the aversive response to ethanol.


Subject(s)
Behavior, Animal/drug effects , Ethanol/pharmacology , Receptors, Glycine/metabolism , Animals , Avoidance Learning/drug effects , Brain/metabolism , Drinking Behavior/drug effects , Food Preferences/drug effects , Mice, Knockout , Motor Activity/drug effects , Mutation , RNA, Messenger/metabolism , Receptors, Glycine/genetics , Reflex, Startle/drug effects
7.
Alcohol Clin Exp Res ; 39(1): 136-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25516156

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor (PPAR) agonists reduce voluntary ethanol (EtOH) consumption in rat models and are promising therapeutics in the treatment for drug addictions. We studied the effects of different classes of PPAR agonists on chronic EtOH intake and preference in mice with a genetic predisposition for high alcohol consumption and then examined human genomewide association data for polymorphisms in PPAR genes in alcohol-dependent subjects. METHODS: Two different behavioral tests were used to measure intake of 15% EtOH in C57BL/6J male mice: 24-hour 2-bottle choice and limited access (3-hour) 2-bottle choice, drinking in the dark. We measured the effects of pioglitazone (10 and 30 mg/kg), fenofibrate (50 and 150 mg/kg), GW0742 (10 mg/kg), tesaglitazar (1.5 mg/kg), and bezafibrate (25 and 75 mg/kg) on EtOH intake and preference. Fenofibric acid, the active metabolite of fenofibrate, was quantified in mouse plasma, liver, and brain by liquid chromatography tandem mass spectrometry. Data from a human genome-wide association study (GWAS) completed in the Collaborative Study on the Genetics of Alcoholism (COGA) were then used to analyze the association of single nucleotide polymorphisms (SNPs) in different PPAR genes (PPARA, PPARD, PPARG, and PPARGC1A) with 2 phenotypes: DSM-IV alcohol dependence (AD) and the DSM-IV criterion of withdrawal. RESULTS: Activation of 2 isoforms of PPARs, α and γ, reduced EtOH intake and preference in the 2 different consumption tests in mice. However, a selective PPARδ agonist or a pan agonist for all 3 PPAR isoforms did not decrease EtOH consumption. Fenofibric acid, the active metabolite of the PPARα agonist fenofibrate, was detected in liver, plasma, and brain after 1 or 8 days of oral treatment. The GWAS from COGA supported an association of SNPs in PPARA and PPARG with alcohol withdrawal and PPARGC1A with AD but found no association for PPARD with either phenotype. CONCLUSIONS: We provide convergent evidence using both mouse and human data for specific PPARs in alcohol action. Reduced EtOH intake in mice and the genetic association between AD or withdrawal in humans highlight the potential for repurposing FDA-approved PPARα or PPARγ agonists for the treatment of AD.


Subject(s)
Alcohol Drinking/genetics , Alcoholism/genetics , PPAR alpha/genetics , PPAR gamma/genetics , Adult , Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Alkanesulfonates/therapeutic use , Animals , Bezafibrate/therapeutic use , Brain/metabolism , Female , Fenofibrate/blood , Fenofibrate/pharmacokinetics , Fenofibrate/therapeutic use , Genome-Wide Association Study , Humans , Liver/metabolism , Male , Mice , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/therapeutic use , Pioglitazone , Polymorphism, Single Nucleotide/genetics , Thiazoles/therapeutic use , Thiazolidinediones/therapeutic use
8.
Front Neurosci ; 8: 129, 2014.
Article in English | MEDLINE | ID: mdl-24904269

ABSTRACT

Some anti-inflammatory medications reduce alcohol consumption in rodent models. Inhibition of phosphodiesterases (PDE) increases cAMP and reduces inflammatory signaling. Rolipram, an inhibitor of PDE4, markedly reduced ethanol intake and preference in mice and reduced ethanol seeking and consumption in alcohol-preferring fawn-hooded rats (Hu et al., 2011; Wen et al., 2012). To determine if these effects were specific for PDE4, we compared nine PDE inhibitors with different subtype selectivity: propentofylline (nonspecific), vinpocetine (PDE1), olprinone, milrinone (PDE3), zaprinast (PDE5), rolipram, mesopram, piclamilast, and CDP840 (PDE4). Alcohol intake was measured in C57BL/6J male mice using 24-h two-bottle choice and two-bottle choice with limited (3-h) access to alcohol. Only the selective PDE4 inhibitors reduced ethanol intake and preference in the 24-h two-bottle choice test. For rolipram, piclamilast, and CDP840, this effect was observed after the first 6 h but not after the next 18 h. Mesopram, however, produced a long-lasting reduction of ethanol intake and preference. In the limited access test, rolipram, piclamilast, and mesopram reduced ethanol consumption and total fluid intake and did not change preference for ethanol, whereas CDP840 reduced both consumption and preference without altering total fluid intake. Our results provide novel evidence for a selective role of PDE4 in regulating ethanol drinking in mice. We suggest that inhibition of PDE4 may be an unexplored target for medication development to reduce excessive alcohol consumption.

9.
PLoS One ; 9(1): e85525, 2014.
Article in English | MEDLINE | ID: mdl-24454882

ABSTRACT

GABAA receptors consisting of ρ1, ρ2, or ρ3 subunits in homo- or hetero-pentamers have been studied mainly in retina but are detected in many brain regions. Receptors formed from ρ1 are inhibited by low ethanol concentrations, and family-based association analyses have linked ρ subunit genes with alcohol dependence. We determined if genetic deletion of ρ1 in mice altered in vivo ethanol effects. Null mutant male mice showed reduced ethanol consumption and preference in a two-bottle choice test with no differences in preference for saccharin or quinine. Null mutant mice of both sexes demonstrated longer duration of ethanol-induced loss of righting reflex (LORR), and males were more sensitive to ethanol-induced motor sedation. In contrast, ρ1 null mice showed faster recovery from acute motor incoordination produced by ethanol. Null mutant females were less sensitive to ethanol-induced development of conditioned taste aversion. Measurement of mRNA levels in cerebellum showed that deletion of ρ1 did not change expression of ρ2, α2, or α6 GABAA receptor subunits. (S)-4-amino-cyclopent-1-enyl butylphosphinic acid ("ρ1" antagonist), when administered to wild type mice, mimicked the changes that ethanol induced in ρ1 null mice (LORR and rotarod tests), but the ρ1 antagonist did not produce these effects in ρ1 null mice. In contrast, (R)-4-amino-cyclopent-1-enyl butylphosphinic acid ("ρ2" antagonist) did not change ethanol actions in wild type but produced effects in mice lacking ρ1 that were opposite of the effects of deleting (or inhibiting) ρ1. These results suggest that ρ1 has a predominant role in two in vivo effects of ethanol, and a role for ρ2 may be revealed when ρ1 is deleted. We also found that ethanol produces similar inhibition of function of recombinant ρ1 and ρ2 receptors. These data indicate that ethanol action on GABAA receptors containing ρ1/ρ2 subunits may be important for specific effects of ethanol in vivo.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, GABA-A/genetics , Animals , Anxiety/psychology , Cells, Cultured , Central Nervous System Depressants/metabolism , Ethanol/metabolism , Female , GABA Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Ketamine/pharmacology , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Phosphinic Acids/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Reflex, Righting/drug effects , Reflex, Startle/drug effects , Rotarod Performance Test , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
10.
J Pharmacol Exp Ther ; 344(2): 489-500, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230213

ABSTRACT

Ethanol is a widely used drug, yet an understanding of its sites and mechanisms of action remains incomplete. Among the protein targets of ethanol are glycine receptors (GlyRs), which are potentiated by millimolar concentrations of ethanol. In addition, zinc ions also modulate GlyR function, and recent evidence suggests that physiologic concentrations of zinc enhance ethanol potentiation of GlyRs. Here, we first built a homology model of a zinc-bound GlyR using the D80 position as a coordination site for a zinc ion. Next, we investigated in vitro the effects of zinc on ethanol action at recombinant wild-type (WT) and mutant α1 GlyRs containing the D80A substitution, which eliminates zinc potentiation. At D80A GlyRs, the effects of 50 and 200 mM ethanol were reduced as compared with WT receptors. Also, in contrast to what was seen with WT GlyRs, neither adding nor chelating zinc changed the magnitude of ethanol enhancement of mutant D80A receptors. Next, we evaluated the in vivo effects of the D80A substitution by using heterozygous Glra1(D80A) knock-in (KI) mice. The KI mice showed decreased ethanol consumption and preference, and they displayed increased startle responses compared with their WT littermates. Other behavioral tests, including ethanol-induced motor incoordination and strychnine-induced convulsions, revealed no differences between the KI and WT mice. Together, our findings indicate that zinc is critical in determining the effects of ethanol at GlyRs and suggest that zinc binding at the D80 position may be important for mediating some of the behavioral effects of ethanol action at GlyRs.


Subject(s)
Alcohol Drinking , Behavior, Animal/drug effects , Ethanol/pharmacology , Point Mutation , Receptors, Glycine/genetics , Zinc/metabolism , Acoustic Stimulation , Alcohol Drinking/genetics , Amino Acid Substitution , Animals , Binding Sites , Electrophysiological Phenomena , Ethanol/administration & dosage , Female , Gene Knock-In Techniques , Homozygote , Male , Mice , Mice, Mutant Strains , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/metabolism , Reflex, Righting/drug effects , Reflex, Righting/genetics , Reflex, Startle/drug effects , Reflex, Startle/genetics , Rotarod Performance Test , Strychnine/pharmacology , Transfection , Xenopus laevis , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...