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 Pharmacol Exp Ther ; 380(3): 162-170, 2022 03.
Article in English | MEDLINE | ID: mdl-35058349

ABSTRACT

The novel wound-healing biologic EPICERTIN, a recombinant analog of cholera toxin B subunit, is in early development for the management of ulcerative colitis. This study established for the first time the pharmacokinetics (PK), bioavailability (BA), and acute safety of EPICERTIN in healthy and dextran sodium sulfate-induced colitic mice and healthy rats. For PK and BA assessments, single administrations of various concentrations of EPICERTIN were given intravenously or intrarectally to healthy and colitic C57BL/6 mice and to healthy Sprague-Dawley rats. After intravenous administration to healthy animals, the drug's plasma half-life (t 1/2) for males and females was 0.26 and 0.3 hours in mice and 19.4 and 14.5 hours in rats, respectively. After intrarectal administration, drug was detected at very low levels in only four samples of mouse plasma, with no correlation to colon epithelial integrity. No drug was detected in rat plasma. A single intrarectal dose of 0.1 µM (0.6 µg/mouse) EPICERTIN significantly facilitated the healing of damaged colonic epithelium as determined by disease activity index and histopathological scoring, whereas 10-fold higher or lower concentrations showed no effect. For acute toxicity evaluation, healthy rats were given a single intrarectal administration of various doses of EPICERTIN with sacrifice on Day 8, recording body weight, morbidity, mortality, clinical pathology, and gross necropsy observations. There were no drug-related effects of toxicological significance. The no observed adverse effect level (intrarectal) in rats was determined to be 5 µM (307 µg/animal, or 5.2 µg drug/cm2 of colorectal surface area), which is 14 times the anticipated intrarectally delivered clinical dose. SIGNIFICANCE STATEMENT: EPICERTIN is a candidate wound-healing biologic for the management of ulcerative colitis. This study determined for the first time the intravenous and intrarectal pharmacokinetics and bioavailability of the drug in healthy and colitic mice and healthy rats, and its acute safety in a dose-escalation study in rats. An initial therapeutic dose in colitic mice was also established. EPICERTIN delivered intrarectally was minimally absorbed systemically, was well tolerated, and induced epithelial wound healing topically at a low dose.


Subject(s)
Biological Products , Colitis, Ulcerative , Wound Healing , Administration, Topical , Animals , Biological Products/administration & dosage , Biological Products/adverse effects , Biological Products/pharmacokinetics , Colitis, Ulcerative/drug therapy , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Rodentia , Wound Healing/drug effects
2.
Microorganisms ; 9(3)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801811

ABSTRACT

Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.

3.
Behav Pharmacol ; 25(2): 147-57, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24603339

ABSTRACT

Binge eating palatable foods has been shown to have behavioral and neurochemical similarities to drug addiction. GS 455534 is a highly selective reversible aldehyde dehydrogenase 2 inhibitor that has been shown to reduce alcohol and cocaine intake in rats. Given the overlaps between binge eating and drug abuse, we examined the effects of GS 455534 on binge eating and subsequent dopamine release. Sprague-Dawley rats were maintained on a sugar (experiment 1) or fat (experiment 2) binge eating diet. After 25 days, GS 455534 was administered at 7.5 and 15 mg/kg by an intraperitoneal injection, and food intake was monitored. In experiment 3, rats with cannulae aimed at the nucleus accumbens shell were maintained on the binge sugar diet for 25 days. Microdialysis was performed, during which GS 455534 15 mg/kg was administered, and sugar was available. Dialysate samples were analyzed to determine extracellular levels of dopamine. In experiment 1, GS 455534 selectively decreased sugar intake food was made available in the Binge Sugar group but not the Ad libitum Sugar group, with no effect on chow intake. In experiment 2, GS 455534 decreased fat intake in the Binge Fat group, but not the Ad libitum Fat group, however, it also reduced chow intake. In experiment 3, GS 455534 attenuated accumbens dopamine release by almost 50% in binge eating rats compared with the vehicle injection. The findings suggest that selective reversible aldehyde dehydrogenase 2 inhibitors may have the therapeutic potential to reduce binge eating of palatable foods in clinical populations.


Subject(s)
Bulimia/drug therapy , Dopamine/metabolism , Enzyme Inhibitors/pharmacology , Isoflavones/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase, Mitochondrial , Animals , Appetite Regulation/drug effects , Appetite Regulation/physiology , Body Weight/drug effects , Bulimia/metabolism , Dietary Fats , Dietary Sucrose , Dose-Response Relationship, Drug , Eating/drug effects , Male , Mitochondrial Proteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
4.
Recent Pat CNS Drug Discov ; 7(2): 151-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22574676

ABSTRACT

Alcoholism is a complex heterogeneous disease and a number of neurotransmitter and neuromodulator systems have been implicated in its manifestation. Consequently, it is unlikely that existing medications such as disulfiram (Antabuse®), naltrexone (ReVia®), acamprosate (Campral®)) can be efficacious in every individual. Thus, the development of novel therapeutic agents with greater selectivity and less unwanted effects for the treatment of this disease is one of the major objectives of alcohol research. This review summarizes the findings of five novel compounds with different neuronal targets for treating alcoholism. These compounds include sazetidine-A, which selectively desensitizes α4ß2 nicotinic receptors; carisbamate, a novel anti-epileptic agent; JNJ5234801, a novel anxiolytic agent; GS-455534, a highly selective inhibitor of mitochondrial aldehyde dehydrogenase; and JNJ-39220675, a selective histamine H3 antagonist. Inbred alcohol-preferring rats (iP), Fawn-Hooded (FH) rats, and P rats were used to evaluate the compounds. Naltrexone was used as a positive control in some experiments. All five compounds reduced alcohol consumption and preference. The mechanisms thought to underlie these effects suggest that, in addition to dopaminergic and opioidergic systems, other neuronal systems such as sodium channels (carisbamate), mitochondrial aldehyde dehydrogenase (GS-455534), 5-HT2 receptors (JNJ-5234801), histamine H3 receptors (JNJ-39220675), and α4ß2 nicotinic receptors (sazetidine-A) can be involved in alcohol drinking. Further work is necessary to confirm the exact mechanisms of action of each drug and to determine any viable targets for putative treatment of alcohol-use disorders. The article presents some promising patents on novel medication targets for the treatment of alcoholism.


Subject(s)
Alcohol Deterrents/pharmacology , Alcoholism/drug therapy , Drug Evaluation, Preclinical/psychology , Molecular Targeted Therapy/methods , Alcohol Deterrents/therapeutic use , Animals , Azepines/pharmacology , Azetidines/pharmacology , Carbamates/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drugs, Investigational/pharmacology , Humans , Isoflavones/pharmacology , Patents as Topic , Piperidines/pharmacology , Pyridines/pharmacology , Synaptic Transmission/drug effects
5.
Nat Med ; 16(9): 1024-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20729865

ABSTRACT

There is no effective treatment for cocaine addiction despite extensive knowledge of the neurobiology of drug addiction. Here we show that a selective aldehyde dehydrogenase-2 (ALDH-2) inhibitor, ALDH2i, suppresses cocaine self-administration in rats and prevents cocaine- or cue-induced reinstatement in a rat model of cocaine relapse-like behavior. We also identify a molecular mechanism by which ALDH-2 inhibition reduces cocaine-seeking behavior: increases in tetrahydropapaveroline (THP) formation due to inhibition of ALDH-2 decrease cocaine-stimulated dopamine production and release in vitro and in vivo. Cocaine increases extracellular dopamine concentration, which activates dopamine D2 autoreceptors to stimulate cAMP-dependent protein kinase A (PKA) and protein kinase C (PKC) in primary ventral tegmental area (VTA) neurons. PKA and PKC phosphorylate and activate tyrosine hydroxylase, further increasing dopamine synthesis in a positive-feedback loop. Monoamine oxidase converts dopamine to 3,4-dihydroxyphenylacetaldehyde (DOPAL), a substrate for ALDH-2. Inhibition of ALDH-2 enables DOPAL to condense with dopamine to form THP in VTA neurons. THP selectively inhibits phosphorylated (activated) tyrosine hydroxylase to reduce dopamine production via negative-feedback signaling. Reducing cocaine- and craving-associated increases in dopamine release seems to account for the effectiveness of ALDH2i in suppressing cocaine-seeking behavior. Selective inhibition of ALDH-2 may have therapeutic potential for treating human cocaine addiction and preventing relapse.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/therapeutic use , Berberine Alkaloids/metabolism , Cocaine-Related Disorders/prevention & control , Dopamine Antagonists/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/therapeutic use , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cocaine/administration & dosage , Cues , Disease Models, Animal , Dopamine/biosynthesis , Enzyme Activation , Infusions, Intravenous , Rats , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism
6.
Alcohol Clin Exp Res ; 33(11): 1935-44, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19673742

ABSTRACT

BACKGROUND: Inherited human aldehyde dehydrogenase 2 (ALDH-2) deficiency reduces the risk for alcoholism. Kudzu plants and extracts have been used for 1,000 years in traditional Chinese medicine to treat alcoholism. Kudzu contains daidzin, which inhibits ALDH-2 and suppresses heavy drinking in rodents. Decreased drinking due to ALDH-2 inhibition is attributed to aversive properties of acetaldehyde accumulated during alcohol consumption. However, daidzin can reduce drinking in some rodents without necessarily increasing acetaldehyde. Therefore, a selective ALDH-2 inhibitor might affect other metabolic factors involved in regulating drinking. METHODS: Aldehyde dehydrogenase 2 inhibitors were synthesized based on the co-crystal structure of ALDH-2 and daidzin. We tested the efficacy of a highly selective reversible ALDH-2 inhibitor, CVT-10216, in models of moderate and high alcohol drinking rats. We studied 2-bottle choice and deprivation-induced drinking paradigms in Fawn Hooded (FH) rats, operant self-administration in Long Evans (LE), FH, and inbred P (iP) rats and in cue-induced reinstatement in iP rats. We also assayed blood acetaldehyde levels as well as dopamine (DA) release in the nucleus accumbens (NAc) and tested possible rewarding/aversive effects of the inhibitor in a conditioned place preference (CPP) paradigm. RESULTS: CVT-10216 increases acetaldehyde after alcohol gavage and inhibits 2-bottle choice alcohol intake in heavy drinking rodents, including deprivation-induced drinking. Moreover, CVT-10216 also prevents operant self-administration and eliminates cue-induced reinstatement of alcohol seeking even when alcohol is not available (i.e., no acetaldehyde). Alcohol stimulates DA release in the NAc, which is thought to contribute to increased drinking and relapse in alcoholism. CVT-10216 prevents alcohol-induced increases in NAc DA without changing basal levels. CVT-10216 does not show rewarding or aversive properties in the CPP paradigm at therapeutic doses. CONCLUSION: Our findings suggest that selective reversible ALDH-2 inhibitors may have therapeutic potential to reduce excessive drinking and to suppress relapse in abstinent alcoholics.


Subject(s)
Alcohol Deterrents , Alcohol Drinking/psychology , Aldehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Isoflavones/pharmacology , Mitochondrial Proteins/antagonists & inhibitors , Acetaldehyde/blood , Aldehyde Dehydrogenase, Mitochondrial , Animals , Choice Behavior/drug effects , Conditioning, Operant/drug effects , Cues , Dopamine/physiology , Extinction, Psychological/drug effects , Male , Microdialysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Pueraria/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recurrence , Self Administration
7.
Alcohol Clin Exp Res ; 28(9): 1308-16, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15365300

ABSTRACT

BACKGROUND: Recent findings suggest that adenosine is involved in the neural and behavioral effects of ethanol (EtOH). Studies in neural cell culture show that EtOH, via activation of adenosine A2 receptors, triggers cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling and CRE (cAMP regulatory element)-mediated gene expression and that this effect is blocked by inhibiting G-protein betagamma subunits. Recently, we reported that expression of a betagamma inhibitor in the nucleus accumbens (NAc) reduces EtOH drinking in rats. The NAc expresses high levels of the adenosine A2A receptor in GABAergic medium spiny neurons. If the reinforcing effects of EtOH are mediated through an A2 activation of cAMP/PKA signaling via betagamma, then A2 receptor blockade should attenuate EtOH consumption. Here we tested this hypothesis. Because adenosine A2 and dopamine D2 receptors are coexpressed in neurons of the NAc, we compared the effects of A2 blockade with those of D2 receptor blockade. METHODS: Male Long-Evans rats were trained to self-administer 10% EtOH in daily 30-min sessions with an active and an inactive lever. Separate groups of rats were given the D2 antagonist eticlopride (0.005, 0.007, and 0.01 mg/kg), the A2 antagonist 3,7-dimethyl-1-propargylxanthine (DMPX; 1, 3, 5, 7, 10, and 20 mg/kg), and the A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX; 0.125, 0.25, and 0.5 mg/kg) by systemic injection. RESULTS: Eticlopride dose-dependently reduced EtOH drinking. DMPX showed a bimodal effect: 10 and 20 mg/kg decreased, but 1 mg/kg increased, EtOH consumption. DPCPX was without effect. CONCLUSIONS: In support of our hypothesis, the A2 antagonist DMPX attenuated EtOH self-administration. Low doses of the A2 antagonist enhanced EtOH drinking, consistent with the possibility that rats increase EtOH self-administration to overcome partial A2 blockade. The D2 antagonist eticlopride also decreased EtOH self-administration. These data provide the first evidence that pharmacological modulation of adenosine A2 receptors can regulate EtOH consumption in rats.


Subject(s)
Conditioning, Operant/drug effects , Ethanol/administration & dosage , Receptors, Adenosine A2/physiology , Theobromine/analogs & derivatives , Adenosine A2 Receptor Antagonists , Animals , Conditioning, Operant/physiology , Dose-Response Relationship, Drug , Male , Rats , Rats, Long-Evans , Self Administration , Theobromine/pharmacology
8.
Cell ; 109(6): 733-43, 2002 Jun 14.
Article in English | MEDLINE | ID: mdl-12086672

ABSTRACT

Dopamine release is activated by ethanol and addicting drugs, but molecular mechanisms linking dopaminergic signaling to neuronal responses and drinking behavior are poorly understood. We report that dopamine-D2 receptors induce PKA Calpha translocation and increase CRE-regulated gene expression. Ethanol also activates PKA signaling. Subthreshold concentrations of the D2 agonist NPA and ethanol, without effect alone, together cause synergistic PKA translocation and CRE-mediated gene transcription. D2 or adenosine A2 receptor blockade, pertussis toxin, Rp-cAMPS, or overexpression of dominant-negative peptides that sequester betagamma dimers prevent synergy. Importantly, overexpression of a betagamma inhibitor peptide in the nucleus accumbens strikingly reduces sustained alcohol consumption. We propose that synergy of D2 and A2 confers ethanol hypersensitivity and that betagamma dimers are required for voluntary drinking.


Subject(s)
Apomorphine/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/metabolism , Ethanol/pharmacology , Receptors, Dopamine D2/metabolism , Receptors, Purinergic P1/metabolism , Adenoviridae/genetics , Alcohol Drinking , Animals , Animals, Newborn , Apomorphine/pharmacology , Blotting, Western , Cyclic AMP/metabolism , Dimerization , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , G1 Phase , Gene Expression Regulation , Genes, Reporter , Hippocampus/cytology , Immunohistochemistry , Integrases/metabolism , Isoenzymes/metabolism , Luciferases/metabolism , Microscopy, Confocal , Models, Biological , Peptides/chemistry , Pertussis Toxin , Protein Binding , Protein Kinase C/metabolism , Protein Kinase C-alpha , Protein Structure, Tertiary , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/chemistry , Receptors, Purinergic P1/chemistry , Signal Transduction , Subcellular Fractions , Time Factors , Transcription, Genetic , Transfection , Viral Proteins/metabolism , Virulence Factors, Bordetella/pharmacology
9.
Acta physiol. pharmacol. ther. latinoam ; 44(3): 70-6, 1994. tab, graf
Article in English | LILACS | ID: lil-152651

ABSTRACT

Se estudió la capacidad de aprendizaje de ratas deprivadas de proteínas durante la etapa perinatal (ratas D) comparándolas con ratas controles (ratas-C) en el test de navegación espacial de Morris. Las ratas-D mostraron mayores latencias de escape para localizar una plataforma sumergida en ausencia de pistas proximales durante el período de adquisición de la prueba. Una experiencia de pre-entrenamiento a las condiciones de nado no mejoró este déficit. Al evaluar la retención de la información espacial a 1, 3, 10 y 30 días posteriores al entrenamiento, no se observaron diferencias significativas entre los grupos. Tampoco se encontró diferencia en el aprendizaje con plataforma visible, resultado que nos permite descartar alteraciones en la capacidad motora del animal deprivado. En conclusión, los resultados sugieren que la hiponutrición perinatal induce un déficit en la capacidad del animal adulto recuperado para resolver eficientemente el test de navegación espacial, aún después de un prolongado período de recuperación nutricional


Subject(s)
Animals , Female , Rats , Pregnancy , Protein Deficiency/diet therapy , Maze Learning , Analysis of Variance , Memory , Rats, Wistar , Reaction Time , Spatial Behavior , Swimming
10.
Acta physiol. pharmacol. ther. latinoam ; 44(3): 70-6, 1994. tab, graf
Article in English | BINACIS | ID: bin-23592

ABSTRACT

Se estudió la capacidad de aprendizaje de ratas deprivadas de proteínas durante la etapa perinatal (ratas D) comparándolas con ratas controles (ratas-C) en el test de navegación espacial de Morris. Las ratas-D mostraron mayores latencias de escape para localizar una plataforma sumergida en ausencia de pistas proximales durante el período de adquisición de la prueba. Una experiencia de pre-entrenamiento a las condiciones de nado no mejoró este déficit. Al evaluar la retención de la información espacial a 1, 3, 10 y 30 días posteriores al entrenamiento, no se observaron diferencias significativas entre los grupos. Tampoco se encontró diferencia en el aprendizaje con plataforma visible, resultado que nos permite descartar alteraciones en la capacidad motora del animal deprivado. En conclusión, los resultados sugieren que la hiponutrición perinatal induce un déficit en la capacidad del animal adulto recuperado para resolver eficientemente el test de navegación espacial, aún después de un prolongado período de recuperación nutricional (AU)


Subject(s)
Comparative Study , Animals , Female , Rats , Pregnancy , Protein Deficiency/diet therapy , Maze Learning , Spatial Behavior , Memory , Swimming , Reaction Time , Analysis of Variance , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...