Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mark J. Benvenga is active.

Publication


Featured researches published by Mark J. Benvenga.


Journal of Pharmacology and Experimental Therapeutics | 2011

N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC), a Novel Metabotropic Glutamate 2 Potentiator with Potential Anxiolytic/Antidepressant Properties: In Vivo Profiling Suggests a Link between Behavioral and Central Nervous System Neurochemical Changes

Matthew J. Fell; Julie F. Falcone; Jason Katner; Kenneth W. Perry; John Hart; Linda Rorick-Kehn; Carl D. Overshiner; Kurt Rasmussen; Stephen F. Chaney; Mark J. Benvenga; Xia Li; Deanna L Marlow; Linda K. Thompson; Susan K Luecke; Keith A. Wafford; Wesley F. Seidel; Dale M. Edgar; Anne T Quets; Christian C. Felder; Xushan Wang; Beverly A. Heinz; Alexander Nikolayev; Ming-Shang Kuo; Daniel Ray Mayhugh; Albert Khilevich; Deyi Zhang; Phillip J Ebert; James E Eckstein; Bradley L. Ackermann; Steven Swanson

The normalization of excessive glutamatergic neurotransmission through the activation of metabotropic glutamate 2 (mGlu2) receptors may have therapeutic potential in a variety of psychiatric disorders, including anxiety/depression and schizophrenia. Here, we characterize the pharmacological properties of N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC), a structurally novel, potent, and selective allosteric potentiator of human and rat mGlu2 receptors (EC50 = 23 and 13 nM, respectively). THIIC produced anxiolytic-like efficacy in the rat stress-induced hyperthermia assay and the mouse stress-induced elevation of cerebellar cGMP and marble-burying assays. THIIC also produced robust activity in three assays that detect antidepressant-like activity, including the mouse forced-swim test, the rat differential reinforcement of low rate 72-s assay, and the rat dominant-submissive test, with a maximal response similar to that of imipramine. Effects of THIIC in the forced-swim test and marble burying were deleted in mGlu2 receptor null mice. Analysis of sleep electroencephalogram (EEG) showed that THIIC had a sleep-promoting profile with increased non-rapid eye movement (REM) and decreased REM sleep. THIIC also decreased the dark phase increase in extracellular histamine in the medial prefrontal cortex and decreased levels of the histamine metabolite tele-methylhistamine (t-MeHA) in rat cerebrospinal fluid. Collectively, these results indicate that the novel mGlu2-positive allosteric modulator THIIC has robust activity in models used to predict anxiolytic/antidepressant efficacy, substantiating, at least with this molecule, differentiation in the biological impact of mGlu2 potentiation versus mGlu2/3 orthosteric agonism. In addition, we provide evidence that sleep EEG and CSF t-MeHA might function as viable biomarker approaches to facilitate the translational development of THIIC and other mGlu2 potentiators.


Journal of Pharmacology and Experimental Therapeutics | 2008

The 5-Hydroxytryptamine2A Receptor Antagonist R-(+)-α-(2,3-Dimethoxyphenyl)-1-[2-(4-fluorophenyl)ethyl-4-piperidinemethanol (M100907) Attenuates Impulsivity after Both Drug-Induced Disruption (Dizocilpine) and Enhancement (Antidepressant Drugs) of Differential-Reinforcement-of-Low-Rate 72-s Behavior in the Rat

Paul A. Ardayfio; Mark J. Benvenga; Stephen F. Chaney; Patrick L. Love; John T. Catlow; Steven Swanson; Gerard J. Marek

Previous work has suggested that N-methyl-d-aspartate (NMDA) receptor antagonism and 5-hydroxytryptamine (5-HT)2A receptor blockade may enhance and attenuate, respectively, certain types of impulsivity mediated by corticothalamostriatal circuits. More specifically, past demonstrations of synergistic “antidepressant-like” effects of a 5-HT2A receptor antagonist and fluoxetine on differential-reinforcement-of-low-rate (DRL) 72-s schedule of operant reinforcement may speak to the role of 5-HT2A receptor blockade with respect to response inhibition as an important prefrontal cortical executive function relating to motor impulsivity. To examine the dynamic range over which 5-HT2A receptor blockade may exert effects on impulsivity, [R-(+)-α-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenyl)ethyl-4-piperidinemethanol] (M100907) was examined both alone and in combination with the psychotomimetic NMDA receptor antagonist dizocilpine [e.g., (-)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate; MK-801] and two different antidepressants, the tricyclic antidepressant desmethylimipramine (DMI) and the monoamine oxidase inhibitor tranylcypromine in rats performing under a DRL 72-s schedule. MK-801 increased the response rate, decreased the number of reinforcers obtained, and exerted a leftward shift in the inter-response time (IRT) distribution as expected. A dose of M100907 that exerted minimal effect on DRL behavior by itself attenuated the psychotomimetic effects of MK-801. Extending previous M100907-fluoxetine observations, addition of a minimally active dose of M100907 to low doses of DMI and tranylcypromine enhanced the antidepressant-like effect of the antidepressants. Therefore, it may be that a tonic excitation of 5-HT2A receptors modulates impulsivity and function of corticothalamostriatal circuits over an extensive dynamic range.


Psychopharmacology | 1995

Olanzapine, an atypical antipsychotic, increases rates of punished responding in pigeons

Mark J. Benvenga; J. David Leander

The effects of olanzapine [LY 170053; 2-methyl-4-(4-methyl-1-piperazinyl)-10H-thieno[2, 3b][1,5]benzodiazepine), a potential atypical antipsychotic, were determined in pigeons whose keypeck responding was punished. These effects were compared to the anxiolytic agents chlordiazepoxide and pentobarbital, and to other antipsychotic agents. Keypeck behavior was maintained under a multiple FR30 FR30 schedule, signalled by white and red stimulus lights, respectively. Each component of the schedule alternated every 3 min with a 30-s timeout. During the white keylight component, responding was maintained by food presentation. During the red keylight component, responding was maintained by food and simultaneously suppressed by electric shock presentation, with response rates being only about 5% of those during the white stimulus light. Olanzapine (0.01–1.0 mg/kg) increased punished responding at doses below those which had an effect on unpunished responding. Clozapine (0.01–1.0 mg/kg), ritanserin (0.1–3.0 mg/kg), and, to a lesser extent, risperidone (0.1–1.0 mg/kg) were also effective at increasing punished responding. Generally, the maximum effect seen with olanzapine was equal to that seen with ritanserin, and it exceeded that seen with clozapine. However, these effects were generally less than those seen with chlordiazepoxide and pentobarbital. Haloperidol (0.01–0.1 mg/kg) was completely without effect on punished responding, while it caused decreases in unpunished behavior. These results provide further evidence that olanzapine has a profile in behavioral tests unlike the typical antipsychotic haloperidol. Moreover, this profile is similar to clozapine, a clinically effective antipsychotic with an atypical profile.


Frontiers in Neuroscience | 2014

LSN2424100: a novel, potent orexin-2 receptor antagonist with selectivity over orexin-1 receptors and activity in an animal model predictive of antidepressant-like efficacy

Thomas E. Fitch; Mark J. Benvenga; Cynthia Darshini Jesudason; Charity Zink; Amy B Vandergriff; Michelle M Menezes; Douglas A. Schober; Linda Rorick-Kehn

We describe a novel, potent and selective orexin-2 (OX2)/hypocretin-2 receptor antagonist with in vivo activity in an animal model predictive of antidepressant-like efficacy. N-biphenyl-2-yl-4-fluoro-N-(1H-imidazol-2-ylmethyl) benzenesulfonamide HCl (LSN2424100) binds with high affinity to recombinant human OX2 receptors (Ki = 4.5 nM), and selectivity over OX1 receptors (Ki = 393 nM). LSN2424100 inhibited OXA-stimulated intracellular calcium release in HEK293 cells expressing human and rat OX2 receptors (Kb = 0.44 and 0.83 nM, respectively) preferentially over cells expressing human and rat OX1 (Kb = 90 and 175 nM, respectively). LSN2424100 exhibits good exposure in Sprague–Dawley rats after IP, but not PO, administration of a 30 mg/kg dose (AUC0–6 h = 1300 and 269 ng*h/mL, respectively). After IP administration in rats and mice, LSN2424100 produces dose-dependent antidepressant-like activity in the delayed-reinforcement of low-rate (DRL) assay, a model predictive of antidepressant-like efficacy. Efficacy in the DRL model was lost in mice lacking OX2, but not OX1 receptors, confirming OX2-specific activity. Importantly, antidepressant-like efficacy of the tricyclic antidepressant, imipramine, was maintained in both OX1 and OX2 receptor knock-out mice. In conclusion, the novel OX2 receptor antagonist, LSN2424100, is a valuable tool compound that can be used to explore the role of OX2 receptor-mediated signaling in mood disorders.


Drug Development Research | 1999

Disinhibitory effects of LY354740, a new mGluR2 agonist, on behaviors suppressed by electric shock in rats and pigeons

Mark J. Benvenga; Carl D. Overshiner; James A. Monn; J. David Leander

Compounds which affect glutamate transmission are reported as anxiolytic in a number of animal models. In the present studies, we evaluated the anxiolytic effect of a new mGluR2 agonist, LY354740, in rats and pigeons. LY354740 was evaluated in three different rat punished responding assays, using different levels of shock intensity. LY354740 and its racemate, LY314582, were generally ineffective at increasing rates of responding suppressed by electric shock. However, both compounds did increase responding during time‐out periods between scheduled components, and both increased responding during a punishment extinction assay. LY354740 was also evaluated in two separate pigeon punished responding procedures. LY354740 was ineffective at increasing punished responding under one schedule, but increased punished responding in the second assay, when shock was not delivered coincident with reinforcement. Clearly, LY354740 was effective in attenuating suppressed responding in some cases, but not all. LY354740 appears to produce a disinhibitory effect on behaviors with low baseline rates of responding (in rats), and a disinhibitory effect on punished behaviors in schedules with less stringent punishment conditions (in rats and pigeons). These results suggest that LY354740 may be useful as a clinically effective anxiolytic. Drug Dev. Res. 47:37–44, 1999.


Drug Development Research | 1997

Increased food consumption by clozapine, but not by olanzapine, in satiated rats

Mark J. Benvenga; J. David Leander

Various drugs used to treat schizophrenia have been repeatedly shown to increase body weight in both animals and humans. There are different theories as to why this occurs, but the most recently studied theory is that these drugs which cause weight gain do so because of an antagonist effect at the 5HT2C receptor. In this work, we studied the effects of olanzapine, clozapine, and risperidone on feeding behavior. Over a 4‐hour test period in satiated rats, clozapine, over a broad dose range, significantly increased food consumption. Similarly, risperidone increased food consumption relative to control. In contrast, olanzapine did not significantly increase food consumption in rats at any dose tested over the 4‐hour test period. This suggests that olanzapine may be different from clozapine and risperidone with respect to potential weight gain in schizophrenic patients. Moreover, we believe that the effect produced by clozapine and risperidone is due to the alpha‐adrenergic activity of these compounds, since olanzapine has a much lower affinity for alpha adrenergic receptors than does clozapine or risperidone, and not due to the 5HT2C activity, which all three compounds have in common. Drug Dev. Res. 41:48–50, 1997.


European Journal of Pharmacology | 1993

Antidepressant-like effect of LY228729 as measured in the rodent forced swim paradigm

Mark J. Benvenga; J. David Leander

The novel 5-HT1A receptor agonist, LY228729, was tested to see if it would produce antidepressant-like behavioural effects in the rodent forced swim model. The tricyclic antidepressant, imipramine, as well as the 5-HT1A receptor agonists, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) and gepirone, produced dose-related decreases in immobility in the forced swim test following subchronic treatment in rats. LY228729, when given over a three injection course, as with the other compounds, significantly reduced immobility at doses of 1 and 3 mg/kg. Subsequent analysis of locomotor activity revealed no increases in behavior, so that general changes in activity could not account for the reduction in immobility time in the forced swim. These results suggest that LY228729 may have clinical antidepressant efficacy.


Drug Development Research | 1997

Pharmacological profile of LY301317, a potent and selective 5-HT1A agonist

Mary C. Wolff; Mark J. Benvenga; David O. Calligaro; Ray W. Fuller; Jaswant Singh Gidda; Susan K. Hemrick-Luecke; James B. Lucot; David L. Nelson; Carl D. Overshiner; J. David Leander

LY301317 ((4r)‐(−)‐4‐(dipropylamino)‐6‐(5‐oxazolinyl)‐1,3,4,5‐tetrahydrobenz[c,d]indole) has high affinity for the 5‐HT1A receptor and weak affinity for the 5‐HT1D and histamine‐H1 receptors. No significant affinity was found for the other amine receptors studied. In rats, LY301317 produced potent in vivo effects that are characteristic of compounds with agonist activity at the 5‐HT1A receptor, such as an increase in serum corticosterone concentration, a reduction in 5‐HIAA concentration in brain tissue, induction of flat body posture and lower lip retraction (components of a serotonin syndrome), and a decrease of core body temperature. In pigeons trained to discriminate 8‐hydroxy‐2‐(di‐n‐propylamino)‐tetralin (8‐OH‐DPAT) from saline, full generalization to LY301317 was observed. LY301317 increased punished responding in both the pigeon and rat conflict tests for anxiolytic activity. LY301317 reduced immobility in the rat forced swim model used to indicate potential antidepressant activity. In pigeons, LY301317 blocked emesis induced by a chemical (ditolyguanidine), by a 5‐HT3 agonist (m‐(chlorophenyl)‐biguanide), and by an oncolytic agent (cisplatin), as well as vomiting induced by conditioning to environmental stimuli (a model of anticipatory nausea and vomiting). In addition, LY301317 blocked cisplatin‐ and ipecac‐induced vomiting in the dog and motion‐induced emesis in the cat. It was concluded that LY301317 is an orally active, potent, and selective agonist for the 5‐HT1A receptor with potential clinical utility as an anxiolytic, an antidepressant, and a broad‐spectrum antiemetic. Drug Dev. Res. 40:17–34, 1997.


Pharmacology Research & Perspectives | 2016

Preclinical findings predicting efficacy and side‐effect profile of LY2940094, an antagonist of nociceptin receptors

Jeffrey M. Witkin; Linda Rorick-Kehn; Mark J. Benvenga; Benjamin L. Adams; Scott D. Gleason; Karen M. Knitowski; Xia Li; Steven Chaney; Julie F. Falcone; Janice W. Smith; Julie Foss; Kirsti Lloyd; John T. Catlow; David L. McKinzie; Kjell A. Svensson; Vanessa N. Barth; Miguel A. Toledo; Nuria Diaz; Celia Lafuente; Alma Jiménez; Alfonso Benito; Conception Pedregal; Maria Angeles Martinez-Grau; Anke Post; Michael Ansonoff; John E. Pintar; Michael A. Statnick

Nociceptin/Orphanin FQ (N/OFQ) is a 17 amino acid peptide whose receptor is designated ORL1 or nociceptin receptor (NOP). We utilized a potent, selective, and orally bioavailable antagonist with documented engagement with NOP receptors in vivo to assess antidepressant‐ and anxiolytic‐related pharmacological effects of NOP receptor blockade along with measures of cognitive and motor impingement. LY2940094 ([2‐[4‐[(2‐chloro‐4,4‐difluoro‐spiro[5H‐thieno[2,3‐c]pyran‐7,4′‐piperidine]‐1′‐yl)methyl]‐3‐methyl‐pyrazol‐1‐yl]‐3‐pyridyl]methanol) displayed antidepressant‐like behavioral effects in the forced‐swim test in mice, an effect absent in NOP−/− mice. LY2940094 also augmented the behavioral effect of fluoxetine without changing target occupancies (NOP and serotonin reuptake transporter [SERT]). LY2940094 did not have effects under a differential‐reinforcement of low rate schedule. Although anxiolytic‐like effects were not observed in some animal models (conditioned suppression, 4‐plate test, novelty‐suppressed feeding), LY2940094 had effects like that of anxiolytic drugs in three assays: fear‐conditioned freezing in mice, stress‐induced increases in cerebellar cGMP in mice, and stress‐induced hyperthermia in rats. These are the first reports of anxiolytic‐like activity with a systemically viable NOP receptor antagonist. LY2940094 did not disrupt performance in either a 5‐choice serial reaction time or delayed matching‐to‐position assay. LY2940094 was also not an activator or suppressor of locomotion in rodents nor did it induce failures of rotarod performance. These data suggest that LY2940094 has unique antidepressant‐ and anxiolytic‐related pharmacological effects in rodents. Clinical proof of concept data on this molecule in depressed patients have been reported elsewhere.


Frontiers in Pharmacology | 2018

Metabotropic Glutamate2 Receptors Play a Key Role in Modulating Head Twitches Induced by a Serotonergic Hallucinogen in Mice

Mark J. Benvenga; Stephen F. Chaney; Melvyn Baez; Thomas C. Britton; William Joseph Hornback; James A. Monn; Gerard J. Marek

There is substantial evidence that glutamate can modulate the effects of 5-hydroxytryptamine2A (5-HT2A) receptor activation through stimulation of metabotropic glutamate2/3 (mGlu2/3) receptors in the prefrontal cortex. Here we show that constitutive deletion of the mGlu2 gene profoundly attenuates an effect of 5-HT2A receptor activation using the mouse head twitch response (HTR). MGlu2 and mGlu3 receptor knockout (KO) as well as age-matched ICR (CD-1) wild type (WT) mice were administered (±)1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) and observed for head twitch activity. DOI failed to produce significant head twitches in mGlu2 receptor KO mice at a dose 10-fold higher than the peak effective dose in WT or mGlu3 receptor KO mice. In addition, the mGlu2/3 receptor agonist LY379268, and the mGlu2 receptor positive allosteric modulator (PAM) CBiPES, potently blocked the HTR to DOI in WT and mGlu3 receptor KO mice. Conversely, the mGlu2/3 receptor antagonist LY341495 (10 mg/kg) increased the HTR produced by DOI (3 mg/kg) in mGlu3 receptor KO mice. Finally, the mGlu2 receptor potentiator CBiPES was able to attenuate the increase in the HTR produced by LY341495 in mGlu3 receptor KO mice. Taken together, all of these results are consistent with the hypothesis that that DOI-induced head twitches are modulated by mGlu2 receptor activation. These results also are in keeping with a critical autoreceptor function for mGlu2 receptors in the prefrontal cortex with differential effects of acute vs. chronic perturbation (e.g., constitutive mGlu2 receptor KO mice). The robust attenuation of DOI-induced head twitches in the mGlu2 receptor KO mice appears to reflect the critical role of glutamate in ongoing regulation of 5-HT2A receptors in the prefrontal cortex. Future experiments with inducible knockouts for the mGlu2 receptor and/or selective mGlu3 receptor agonists/PAMs/antagonists could provide an important tools in understanding glutamatergic modulation of prefrontal cortical 5-HT2A receptor function.

Collaboration


Dive into the Mark J. Benvenga's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge