Member Spotlight & In the News

Members in the News

Michelle Doyle: MDPV Self-administration in Female Rats – Influence of Reinforcement History By Rachel Altshuler Ph.D., Animals in Research Committee Member Sex differences in addiction research studies are well-noted at the behavioral level, both in humans and animal models of substance use disorders. For example, women progress more rapidly from casual drug use to a substance use disorder. Likewise, cocaine acquisition in rodents tends to be faster in female rats than in male rats. These findings strengthen the argument for including male and female subjects in addiction research studies. While sex differences have been well characterized in some stimulants, sex differences in 3,4-methylenedioxypyrovalerone (MDPV) are not as well characterized. MDPV, a monoamine uptake inhibitor, is part of a family of synthetic cathinones also known as “bath salts.” MDPV and other bath salts are a public health concern due to their high abuse liability and marketing as the cheaper option to stimulants such as cocaine. Interestingly, a “high-responder” phenotype has been observed in subset of male rats that self-administration MDPV, and this phenotype carries over to other stimulants. However, some evidence suggested a history of cocaine self-administration may prevent the development of the phenotype. Understanding whether sex plays into the high-responder phenotype is of particular interest to Michelle Doyle, a Ph.D. student in laboratory of Dr. Greg Collins in the Department of Pharmacology at the University of Texas Health Science Center at San Antonio. In a recent study (Doyle et al. 2021 Psychopharmacology) Michelle Doyle and her colleagues examined whether the high-responder phenotype also developed in female rats self-administering MDPV, as well as, further characterized whether a history of cocaine self-administration protected against the development of this phenotype. Female rats were trained to self-administer MDPV, cocaine, or food pellets. The rats that initially responded for cocaine or food pellets were later switched to MDPV. Rats were classified as high-responders if more than 20% of their responses were made during time-out periods. Dose-response curves for MDPV were generated under a fixed ratio schedule of reinforcement to measure reinforcing potency, then the rats were switched to a progressive ratio (PR) schedule of reinforcement to measure the reinforcing effectiveness of MDPV. Half of the rats self-administering MDPV were characterized as high-responders, whereas only a single rat self-administering cocaine and no rats self-administering food pellets were characterized as high-responders. When all rats were switched to self-administering MDPV, more than half of the rats with a history of responding for food pellets developed the high-responder phenotype. However, no rats with a history of cocaine self-administration developed the high-responder phenotype during MDPV self-administration. There were no differences in the reinforcing potency or effectiveness of MDPV between the rats with different drug histories. This study demonstrated that a subset of female rats develops a high-responder phenotype following MDPV self-administration. Furthermore, protection against the development of the high-responder phenotype cannot be afforded simply to a history of operant behaviors, rather a history of cocaine responding is important for the protective effects. Michelle and colleagues noted that the outcomes of these studies were similar to earlier studies using male rats under the same conditions, suggesting that there may not be sex differences in MDPV self-administration or the development of the high-responder phenotype. Michelle Doyle’s introduction to psychopharmacology began as an undergraduate student at Tufts University, working in the laboratory of Dr. Klaus Miczek. With Dr. Miczek, Michelle first became involved with addiction research when she studied the intersection between stress and cocaine self-administration. She followed her interest in this line of research to the McLean Hospital and the Harvard Medical School where she worked as a technician in the laboratory of Dr. Jack Bergman, under the direction of Dr. Rajeev Desai. Michelle Doyle is entering her final year of her Ph.D. program and will soon be looking for postdoctoral research opportunities. She is particularly interested in studying treatments for substance-use disorders as well as identifying the processes that factor into substance-use disorders. Michelle has been a member-in-training of CPDD since 2018. In addition to many awards won over her time as a graduate student, Michelle was awarded the NIDA Director’s Travel Award and the NIDA Women and Sex/Gender Junior Investigator Travel Award to attend the annual CPDD meetings in 2021 and 2020, respectively. She also won the Excellence in Preclinical Work by a Pre-Doctoral Researcher Award at the Early Career Research Presentation Competition at the 2021 meeting.
Dr. Jacques Nguyen: Behavior and Neural Circuits Underlying Opioid Addiction and Dependence By Sally Huskinson, Ph.D., Animals in Research Committee Member The opioid overdose crisis has received considerable attention since the beginning of the 21st century, and while the current (third-wave) crisis is fueled largely by synthetic opioids like fentanyl, oxycodone is still among the most prescribed opioid analgesics for the treatment of pain. Unfortunately, those who misuse oxycodone or other prescription opioids are at an increased risk of transitioning to illicit opioid use, particularly when their prescription has ended and/or during periods of withdrawal. Surprisingly few preclinical researchers have evaluated neurobiological mechanisms underlying key features of addiction with oxycodone, including escalation of drug intake, the development of physical dependence, and withdrawal. This is one area of focus in the laboratory of Dr. Jacques Nguyen, Ph.D. Currently, Dr. Nguyen is an Assistant Project Scientist in the Department of Psychiatry at the University of California San Diego in La Jolla, California. Dr. Nguyen was recently awarded a K99/R00 Award from the National Institute on Drug Abuse (NIDA) to elucidate behavior and neural circuits underlying opioid addiction and dependence, and this award has already produced important results. In a recent publication (Nguyen et al. 2021, British Journal of Pharmacology), Dr. Nguyen and his colleagues used Wistar rats in intracranial self-stimulation (ICSS), a procedure in which an animal responds on an operant manipulandum (e.g. quarter turn of a wheel) to receive pulses of electrical brain stimulation to a brain region of interest, often in the medial forebrain bundle, and ICSS thresholds can be obtained. When ICSS thresholds are lowered, this reflects a facilitation of brain stimulation reward, and acute administration of drugs of abuse, like oxycodone, lower ICSS thresholds. When ICSS thresholds are elevated, this reflects reduced or diminished reward. Elevated thresholds occur following activation of kappa-opioid receptors and during withdrawal from chronic opioid administration. In addition to their ICSS procedure, Dr. Nguyen and his colleagues used intravenous oxycodone self-administration to investigate potential affective consequences (via ICSS) of escalating opioid intake as well as withdrawal from opioid self-administration. Different groups of male Wistar rats self-administered oxycodone in either short- (1-hour sessions) or long- (11 or 12-hour sessions) access conditions. Subjects completed self-administration sessions five days per week, and this was followed by an intermittent, 60-hour discontinuation of access to oxycodone. As expected, subjects in the long-access group escalated their oxycodone intake while subjects in the short-access group did not escalate their oxycodone intake. Furthermore, escalation of intake increased in a stepwise pattern across successive weeks of oxycodone self-administration beginning with the first day following the 60-hour abstinence period. This latter finding indicates a facilitation of escalation of drug intake after intermittent abstinence, and if translated to humans, suggests that drug taking will be greater after a period of abstinence than the prior drug-taking episode. Moreover, escalated intake was associated with elevated ICSS thresholds, indicative of a diminished reward state and of a growing negative affective state that could be blocked via administration of a kappa-opioid receptor antagonist. Perhaps the most unexpected finding was that elevated ICSS thresholds began to normalize following the 60-hour abstinence period, and this normalization paradoxically facilitated escalation of oxycodone intake once long-access self-administration sessions resumed. In addition, ICSS thresholds could be normalized by providing access to a 1-hour oxycodone self-administration session. Dr. Nguyen also found that pretreatment with the phytocannabinoid, Δ9-tetrahydrocannabinol (THC), reduced oxycodone intake but did not alter the increase in reward threshold that was observed across sequential self-administration sessions, likely illustrating changes in the reinforcing efficacy of a unit dose of oxycodone. From these experiments, Dr. Nguyen and colleagues concluded that changes in brain reward function (i.e., ICSS thresholds) that occur during escalation of oxycodone intake were driven by an interaction between negative affective states and restoration of brain reward status during longer periods of abstinence. These results have clinically meaningful implications for adherence to prescription opioid medications in that early nonadherence could serve to increase prescription opioid intake following the period of nonadherence. Dr. Nguyen completed his doctoral training with his mentors, Drs. Michael Forster and Michael Gatch, in Biomedical Sciences-Pharmacology and Neuroscience at the University of North Texas Health Science Center in Fort Worth, Texas and his postdoctoral training with his mentor, Dr. Michael Taffe, in Neuroscience at the Scripps Research Institute in La Jolla, California. Dr. Nguyen has been a member of CPDD since 2011 and is a regular attendee at the Annual Meeting for CPDD. In addition, Dr. Nguyen was the recipient of the prestigious Stephen G. Holtzman Travel Award for Preclinical Investigators in 2019 and has been a member of the Code of Conduct Working Group for CPDD since 2018 and the CPDD Communications Committee since 2021.
Dr. Michael Taffe: Development of an e-Cigarette Model for the Study of Cannabinoid Vaping in Rats By Lais F. Berro, Ph.D., Animals in Research Committee Member Vaping devices, also known as e-cigarettes, have become increasingly popular, particularly among teenagers. Although initially promoted for cigarette smokers, the e-cigarette technology has also markedly changed marijuana use. Millions of people now vape cannabis and cannabis extracts, with epidemiological studies showing that nearly 10% of teenagers report having vaped cannabis in the past year. Many cannabis e-cigarette users report vaping ∆9‐tetrahydrocannabinol (THC), the ingredient in marijuana that makes people feel “high”. However, the vaping of cannabidiol (CBD), another ingredient of marijuana, has also increased in recent years due to the claimed health benefits of this compound. Anecdotal and empirical evidence suggests that CBD use can improve mood, sleep, anxiety, pain, and even acne. Despite the recent and growing trends in THC and CBD vaping, not enough research has been done to understand the benefits and risks of this form of cannabis use. This gap in scientific knowledge exists not only because the vaping technology is still relatively new, but also due to federal rules that limit research into the effects of cannabis in humans. Therefore, this knowledge gap can only be filled by the employment of animal models. But how to model vaping in animal subjects? Dr. Michael Taffe and his research team at The Scripps Research Institute (the Taffe Laboratory is now at the University of California-San Diego) were up to the task. In the last 5 years, the Taffe laboratory has developed and validated a method for delivering THC and CBD (among other drugs of abuse) to rats using commercial e-cigarette technology. Using this model, which consists of placing the rats in a sealed chamber that regulates airflow and the delivery of vaporized drug using e-cigarette devices, Dr. Taffe and his group have investigated many health and behavioral effects of CBD and THC vaping. Among these findings, studies from Dr. Taffe’s laboratory have shown that THC, but not CBD, inhalation decreased pain sensitivity in rats. These studies point to THC as the main cannabis constituent responsible for its pain-decreasing effects and show that CBD may not be effective for some kinds of pain. Importantly, they also showed that the effects of THC vaping on pain sensitivity were reduced with repeated daily THC inhalation, an effect called “tolerance”. Female rats were more likely to develop tolerance than males. Therefore, repeated THC vaping could make its beneficial effects on pain less robust over time, especially in women. Given the increasing popularity of cannabis extract vaping among teenagers, Dr. Taffe and his team have also set out to investigate the consequences of adolescent THC vaping using their new rat e-cigarette model. They have shown that repeated THC vaping in adolescent rats has lasting consequences into adulthood. After repeated THC vaping as adolescents, adult male rats consume more food, and adult female rats have reduced pain-decreasing effects in response to THC. A particularly alarming finding from this research was that adolescent THC vaping increased the abuse potential of the opioid drug fentanyl in adult female rats. Opioid drugs are potent pain relievers, but because they also produce euphoria, opioid misuse and abuse has increased dramatically in the United States. The US opioid crisis has led to increased restrictions on prescription opioids, with a consequence being the urgent need for alternative pain killers. As an increasing numbers of patients turn to medicinal cannabis to fight pain, they also have used it to enhance the effects of opioid analgesics. Unfortunately, little is known about the safety of this combination. Using their rat “vaping chambers”, Dr. Taffe’s group has looked at whether THC vaping can alter how much adult male rats take of the prescription opioid oxycodone (OxyContin®) in a rat abuse model. Their results were very encouraging: THC vaping not only decreased the intake of oxycodone, but it also potentiated its pain-decreasing effects. Together, these data supported the potential use of THC to enhance the therapeutic efficacy, and to reduce the abuse, of prescription opioids. Going back to their study in adolescent rats, however, the same effects were not observed when THC vaping happened during adolescence. Instead, adolescent THC vaping only increased the intake of the opioid fentanyl in female rats when studied as adults. Therefore, THC vaping during adolescence seems to be particularly dangerous, a critical finding in light of the large number of teenagers who now vape cannabis and cannabis extracts. The work of Dr. Michael Taffe has shed light into many health effects of THC and CBD, both positive and negative. His work in adolescent rats has also highlighted the importance of raising awareness about the potentially dangerous and long-lasting consequences of cannabis extract vaping among teenagers. The establishment of a highly translational rodent e-cigarette model represents a major advancement in cannabinoid science. As recent reports of problems associated with THC vaping increase among human users, Dr. Taffe’s research is at the forefront of investigating the therapeutic and deleterious effects of cannabinoid vaping. We look forward to learning about his future contributions to the field. Michael A. Taffe, Ph.D., has been a member of CPDD since 1998 and has served on the Publications and Media committees. Lais F. Berro, Ph.D., has been a member of CPDD since 2018 and currently serves on the Animals in Research committee.
Dr Jay McLaughlin: Screening Novel Compounds as Analgesics and Therapeutics By Lance McMahon, Ph.D., Animals in Research Committee Member In a time of intense focus on the opioid crisis, some have been committed to providing solutions for decades. Enter Jay McLaughlin, Ph.D., an Associate Professor of Pharmacodynamics at the University of Florida. With a cadre of multidisciplinary collaborators including Lawrence Toll, Siobhan Malany, Susruta Majumdar, Jane Aldrich and Christopher McCurdy, Jay has been intensely focused on analgesic drug discovery and development, including both small and large molecules spanning multiple targets within the CNS and peripheral pain-related pathways. But Jay’s scientific prowess does not stop there, as he also investigates biological mechanisms by which HIV mediates neuropathology and alters behavior, including drug abuse and dependence. Jay’s overarching research goal is to identify the neurobiological systems underlying behavior and psychological disorders, to characterize them, and to develop new therapeutic interventions utilizing molecular, pharmacological, anatomical and behavioral methods. His projects encompass both basic science and drug discovery elements. Screening of novel compounds as analgesics and therapeutics for drug abuse has been central to Dr. McLaughlin’s scientific mission. Continuing a 14-year collaboration, he works with Jane Aldrich at the University of Florida to explore peptide kappa opioid agonists and antagonists as therapeutics for substance abuse. These include a set of cyclic tetrapeptides showing both mixed opioid-receptor agonism and kappa-opioid receptor antagonism, which may have promise as both analgesics and therapeutics for drug abuse. Newer compounds arising from this series show multifunctional opioid activity while preventing relapse to extinguished cocaine-seeking behavior, and in recent tests, morphine-seeking behavior. An additional collaboration seeking novel analgesics is currently underway with Chris McCurdy at the University of Florida. This collaboration has yielded two lines of research. In the first, novel sigma receptor ligands are being characterized for analgesic effects, particularly for alleviation of neuropathic pain, without addictive or sedative effects. In a second project, small molecule guanidine-piperidine agonists and antagonists selective for neuropeptide FF (NPFF)-receptor subtypes are showing promise as an approach to prevent acute morphine antinociceptive tolerance. Particularly intriguing are ligands from this series with dual opioid- and NPFF-receptor activity. The widespread use of combined antiretroviral therapies has worked to curtail the expression and spread of the Human Immunodeficiency Virus (HIV), but neuropathological and behavioral disorders persist that cannot be presently accounted for as mere symptoms of the disease. These deficits, which include motor incoordination, learning and memory disruption, and affective disorders, comprise a syndrome referred to as NeuroAIDS. The HIV regulatory protein, Tat, has been implicated in the pathogenesis of HIV-1 neurological complications, but direct demonstrations of the effects of Tat on behavior and neurodegeneration in an intact organism in vivo are limited. Jay is currently on the front lines working with innovative animal models such as Dr. Johnny He’s iTat mouse. These mice contain a doxycycline (Dox)-inducible and brain-selective tat gene coding for Tat protein, allowing the controlled expression of Tat protein for selective study on desired pathological and behavioral questions. Jay is pioneering the use of iTat mice to test the hypothesis that the activity of Tat in brain is sufficient to produce neuronal dysfunction and neurodegeneration leading to behavioral deficits in learning and memory performance, mood disorders, and increases in drug-seeking behaviors. Dr. McLaughlin’s scientific family tree consists of a who’s who in opioid receptor chemistry, pharmacology and physiology. Jay received PhD training under the mentorship of Jean Bidlack, PhD, Professor of Pharmacology and Physiology at University of Rochester Medical Center, followed by postdoctoral training under Charles Chavkin, PhD, Professor of Pharmacology at the University of Washington. Prior to joining UF, Jay was an Assistant Professor of Psychology at Northeastern University and an Associate Member at the Torrey Pines Institute for Molecular Studies in Port St. Lucie, Florida. Among his numerous talents, Jay has a rapier wit, which is sometimes evidenced by his uncanny knack for explaining life’s deepest complexities by analogy to popular song and film from the 80s. When that doesn’t work, current Netflix series provide reasonable consolation. Dr. McLaughlin first starting attending CPDD as a student in 1995, and has been a member since 2005.
Dr. Matthew Banks: Investigating Novel Treatment Approaches for Opioid Use Disorder in Translational Rodent Models By Paul W. Czoty, Ph.D., Animals in Research Committee Member Deaths due to opioid overdose have risen steadily for nearly two decades. Whereas early phases of this epidemic were largely attributable to heroin and “prescription opioids” such as oxycodone, the past 5 years have seen a rapid increase in deaths due to abuse of other synthetic opioids, such as the highly potent mu opioid receptor agonist fentanyl. With yearly opioid overdose deaths now eclipsing 70,000 per year, novel therapeutic approaches are needed. Laboratory animal models can aid progress in this regard, provided that their results have high translatability to the clinical condition. At Virginia Commonwealth University in Richmond, Dr. Matthew Banks and his colleagues developed a procedure in which rats could choose to receive either an intravenous injection of fentanyl or a food reward. Compared to procedures in which animals have a single response option, choice procedures better reflect the fact that drug abusers choose to allocate their time, resources and behavior towards procurement of drugs rather than other stimuli in the environment such as work, family or education. In previous attempts to study choice behavior in rodents, however, rats typically chose to respond to receive food reinforcement rather than injections of opioids. In the procedure developed by Dr. Banks and colleagues, rats responded almost exclusively for food when the simultaneously available fentanyl dose was low, but chose fentanyl more and more as the fentanyl dose increased, consistent with human laboratory and nonhuman primate choice studies. In the April 2019 issue of Neuropsychopharmacology, Dr. Banks’ group reported results that help to validate the translational utility of this model. Male and female rats chose between injections of fentanyl (0.01-0.1 mg/kg/hour) and access to liquid food. When treated with the mu opioid receptor antagonist naltrexone continuously for 7 days, choice of fentanyl decreased to a degree that was related to naltrexone dose. That the model was sensitive to naltrexone, an FDA-approved treatment for opioid use disorder, supports the contention that results from the model will translate to humans. In further support of this view, the model was sensitive to a negative control, the alpha-2 adrenergic receptor agonist clonidine. When rats were treated for 7 days with clonidine (0.0032-0.01 mg/kg/hour), fentanyl choice was increased. These data are reminiscent of results in humans because, although clonidine can reduce some somatic signs of opioid withdrawal, it has proven ineffective in preventing relapse. Having provided support for the translational utility of this model, Dr. Banks and colleagues then used it to test whether a fentanyl vaccine could reduce fentanyl choice. In non-dependent, vaccinated rats, fentanyl choice decreased and food choice increased over 15 weeks, indicating a highly effective attenuation of the reinforcing effects of fentanyl. In another experiment, the hypothesis that the vaccine would prevent withdrawal-induced increases in fentanyl choice was tested. The ability of opioid withdrawal to increase the reinforcing effects of opioids is well documented in the preclinical and clinical literature. Rats were provided access to fentanyl for 12 hours per day for 14 days under a procedure in which they received a fentanyl injection after every 5 responses. These conditions led to high fentanyl intakes and the development of dependence. When fentanyl-food choice was assessed 8 hours after the end of extended-access sessions, fentanyl choice was decreased and choice of food was increased in vaccinated rats relative to their behavior at the start of the extended-access regimen. The opposite effects (the expected withdrawal-induced increases in fentanyl choice) were observed in non-vaccinated rats. Taken together, the results present compelling evidence of the effectiveness of immunotherapies in combating the ongoing opioid epidemic. Dr. Banks is currently an Associate Professor in the Department of Pharmacology and Toxicology at Virginia Commonwealth University. He received his Pharm.D. degree from Ohio Northern University under the mentorship of Dr. Jon Sprague and received his Ph.D. from Wake Forest University working in the laboratory of Dr. Michael Nader. At both institutions, his research involved the toxic and abuse-related effects of MDMA. He subsequently completed postdoctoral fellowships at Emory University and VCU under the mentorship of Dr. Leonard Howell and Dr. Steve Negus, respectively. Dr. Banks has been regularly attending CPDD since 2004 and joined CPDD as a member-in-training in 2005. He received the CPDD Joseph Cochin Young Investigator Award in 2017. He currently is serving on the CPDD Program committee. Dr. Czoty has participated in CPDD meetings since 1998. He is currently a member of the Animals in Research Committee.
Dr. Kathryn Cunningham: Investigating the Role of Serotonin in Relapse Vulnerability in Cocaine Use Disorder By Dustin J Stairs, Ph.D., Animals in Research Committee Member Cocaine use disorder (CUD) is a disorder with high rates of relapse. Despite these high rates of relapse, there are no current FDA-approved medications to prevent cocaine relapse. Two factors that have been shown to influence rates of relapse is the trait of impulsivity and an individual’s level of reactivity to cocaine-related cues. Understanding the underlying neural mechanisms of how both impulsivity and reactivity to cocaine cues influence cocaine relapse could potentially lead to better pharmacotherapeutic treatment options to enhance recovery from CUD. To investigate this question, Dr. Kathryn Cunningham and her colleagues at the University of Texas Medical Branch in Galveston have used various rodent animal models of both impulsivity and cue reactivity. Through their research, Dr. Cunningham’s team has shown that there is a common serotonergic pathway that underlies the ability of both impulsivity and cue-reactivity to serve as relapse vulnerability factors in CUD. In a recently published study in the Journal of Pharmacology and Experimental Therapeutics, Dr. Cunningham’s group looked specifically at the role of the 5-HT2A receptor in impulsive action and cocaine cue reactivity in rats. To exam this, the team first tested the effects of the FDA-approved, 5-HT2A receptor antagonist pimavanserin on the 1-choice serial reaction time task (1-CSRT) of impulsive action. They obtained baseline impulsive action on the 1-CSRT task in a second group of rats who then were tested for cocaine cue reactivity by allowing them to acquire stable levels of cocaine self-administration followed by forced abstinence for either 1 day or 30 days. Either one day or 30 days after cocaine abstinence, rats were pretreated with various doses of pimavanserin or saline prior to measurement of levels of drug-seeking responses. Dr. Cunningham’s group found that pimavanserin suppressed impulsive action, and that the baseline level of impulsive action predicted the levels of cocaine cue reactivity on day 30 of abstinence. They also found that baseline levels of impulsive action predicted the effectiveness of pimavanserin to decrease the level of cocaine cue reactivity after 30 days of abstinence. These data are particularly exciting as indicating a potential serotonergic drug, which already has FDA approval, could be useful as a treatment for relapse in CUD. Also, the utility of the drug to treat cocaine relapse may be increased if the individual’s level of impulsivity is also considered in the treatment plan. This research conducted in a preclinical, animal model is an important step in the process of medications development for CUD. Given the extensive time and expense required to move novel compounds toward clinical use, animal studies are helpful in optimizing clinical trials with regard to assessment methods and endpoints which serve as indicators of success or failure. Dr. Cunningham has a long history of participation and leadership in CPDD. She won a travel award to attend the 1988 CPDD meeting and became a CPDD member in 1993, and became a fellow in 2007. Dr. Cunningham has chaired the Publications and Nominating Committees, served on the Board of Directors, and was elected CPDD President and member of the Executive Committee. She was awarded the CPDD Mentorship Award (2012) and the Marian W. Fischman Memorial Award (2013). Dr. Stairs has been a member of CPDD since 2010. He currently serves as a member on the Animals in Research Committee.  
The influence of biological sex and gonadal hormones on substance use remains an important area of research in addiction science. Epidemiological data highlight the role that biological sex may play in the development of substance use disorders as well as a resistance to intervention. Animal models help provide important information about the role of these sex differences in drug-taking behavior by isolating the influence of biological sex across varied stages of substance use disorder. New research by Dr. Ryan Lacy at Franklin & Marshall College uses a preclinical model of behavioral economic demand to evaluate the role of biological sex and gonadal hormones on opioid and cocaine use. This study is a part of Dr. Lacy’s broader research program utilizing preclinical models to evaluate the neurobiological and behavioral effects of gonadal sex hormones on drug-seeking and drug-taking behavior. Male and female rats were tested using a within-session threshold procedure to determine behavioral economic demand for cocaine and the short-acting opioid remifentanil. This threshold procedure allows for the rapid evaluation of drug demand by manipulating the “cost” of a substance within a single session. Responses can be modeled using behavioral economic demand functions to determine independent behavioral mechanisms underlying consumption, including use at unconstrained price and price sensitivity. This procedure provides a translationally relevant measure of substance use by evaluating the consumption of a drug under competing environmental costs in a way that can be similarly evaluated in the human laboratory and clinic. Subjects were tested for cocaine and remifentanil demand using this threshold procedure over 15 days in a counterbalanced testing order. Overall, no differences between male and female subjects were observed for cocaine or remifentanil demand. However, among female subjects, a significant effect of freely cycling hormones was observed. Female subjects showed higher levels of unconstrained demand (consumption at low cost) for both cocaine and remifentanil during times in which circulating estrogen was high (i.e., during the estrus phase). These effects were detected by tracking and recording estrous cyclicity throughout behavioral testing using a vaginal lavage procedure. This low-cost and relatively noninvasive procedure is one that any laboratory conducting research with female subjects can effectively and efficiently incorporate into their studies to evaluate gonadal hormones without the need for additional surgical procedures and/or exogenous hormone administration. The counterbalanced testing order of remifentanil and cocaine afforded the opportunity to study exposure effects on drug demand. These analyses indicated a robust effect of prior remifentanil exposure on cocaine demand with increases in cocaine intake at low price and diminished price sensitivity. Exposure effects were pharmacologically specific in that prior cocaine exposure did not impact remifentanil demand. Sex differences were not observed for these exposure effects, consistent with the lack of sex differences in overall drug demand. This study highlights the importance of including male and female subjects in behavioral pharmacology research as well as the utility of using preclinical animal models for evaluating the influence (or lack of influence) of biological sex and gonadal hormones on substance use. Preclinical research historically used only male subjects due to concerns over cycling sex hormones in female subjects. Recently, however, researchers such as Dr. Lacy have seen the inclusion of female subjects not as a limitation, but as an opportunity to improve the generalizability and richness of preclinical research. This body of preclinical work represents an important foundation for understanding the neurobiology of sex differences in drug-taking behaviors to help ensure the effective and comprehensive development of prevention and treatment efforts for substance use disorder. Ryan Lacy has been a member of CPDD since 2008 and currently serves on the Publications committee. Justin Strickland has been a member of CPDD since 2014 and currently serves on the Animals in Research and Travel Award committees. The study was supported by Research Development Funds from Franklin & Marshall College and the National Science Foundation. This study is published in Addiction Biology. By Justin C. Strickland, Ph.D., Animals in Research Committee Member
Opioid and other drug overdoses resulted in more than 70,000 deaths in the US in 2017. While this issue has justifiably garnered a great deal of public attention, it is important not to forget a much more insidious and prolific killer: smoking. Cigarette smoking contributes to almost half a million deaths each year, including 40,000 from passive smoke exposure, alone. Mary Jeanne Kallman has spent much of her career in industry – teaching, managing, and directing research – but nicotine dependence and experimental treatment thereof is an issue that she has managed to revisit repeatedly in her career. Her major contributions to the field include quantitation of nicotine withdrawal and assessment of various experimental treatments of nicotine dependence using preclinical models. Nicotine (or any other drug’s) withdrawal is often measured via application of a functional observational battery – a series of tests which are mandated by regulators. However, the test is often insensitive to subtle withdrawal signs and has components requiring substantial subjective input, and hence, bias. One commonality in drug withdrawal is anxiety, and Mary Jeanne and collaborators made extensive, successful use of acoustic startle to quantify the magnitude of nicotine and other drugs’ dependence properties, and reversal thereof. Like many of us who have behavioral pharmacology labs in industry, when new high specificity and affinity ligands arrive, we like to test them in model systems that we care about, such as drug reinforcement and dependence. As such, Mary Jeanne was the first to demonstrate the potential efficacy of CCK-B antagonists, 5-HT1a antagonists, and mGLUR2 agonists in nicotine dependence. Any of these appear worthy of clinical testing. Mary Jeanne earned a PhD from the University of Georgia in Neuroscience and completed a postdoctoral fellowship in pharmacology and toxicology at Virginia Commonwealth University. She was a tenured Associate Professor and Director of the PhD Experimental Graduate Program at the University of Mississippi for 9 years. She worked for Lilly Research Labs for 17 years as the Group Leader for Safety Pharmacology in their Toxicology Group where she provided drug development solutions in rodents, primates and dogs as needed. She also worked for Covance Laboratories, a large CRO, as the Global Director of Neuroscience and she is currently managing a consulting business which provides input on CNS issue resolution and abuse liability strategies for drugs in development. Mary Jeanne holds diplomate status in the Safety Pharmacology Society and she has held several professional leadership positions including President of the Safety Pharmacology Society, Leader of the Scientific Liaison Coalition (SLC) of SOT, and co-leader of the Cross- Company Abuse Liability Council (CCALC). She frequently publishes scientific papers, book chapters, and presents on various CNS issues. She has been a member of CPDD since 2003. By Thomas Hudzik, Ph.D., Animals in Research Committee Member.
Drug abuse is a multimodal disorder which can be characterized by behavioral, neurological, and pharmacological dysfunction. Multiple lines of preclinical science have explored these facets of addiction while/by utilizing various techniques influenced by various fields of research. However, multidisciplinary approaches to addiction research have begun to develop a more complete and broader understanding of how these various aspects of drug-taking and drug-seeking behavior interact to maintain abuse-related pathologies. Preclinical models of drug-taking behavior (i.e., intravenous self-administration in animals) have been an invaluable tool in evaluating the reinforcing effects of drugs and examining candidate medications for the treatment of substance use disorders. Yet little is known about the neurobiological mechanisms involved not only with drug consumption, but stimuli associated with drug-taking and drug-seeking behavior. In an effort to elucidate the complex interaction between behavior and drug-consumption, Dr. Stephen Kohut, an Assistant Professor of Psychiatry at McLean Hospital/Harvard Medical School, has been developing neuroimaging procedures to examine the effects of drugs in nonhuman primates. These studies can be broadly divided into two main categories: 1) changes in resting state neural connectivity in anesthetized monkeys following repeated drug exposure, and 2) changes in neural connectivity in awake monkeys self-administering drugs. To conduct these experiments, Dr. Kohut designed custom chambers so that monkeys can rest comfortably within the bore of an fMRI. For experiments in awake monkeys, Dr. Kohut constructed topographical maps of each individual monkey’s head to make custom-fitted masks that constrain movement and allow for clean imaging data. Furthermore, Dr. Kohut established a novel fixed ratio operant procedure in which pressing a lever for an established period of time resulted in drug injections. This hold-down fixed ratio procedure limits movement that traditional operant self-administration tasks create. Studies conducted in anesthetized monkeys aimed to elucidate the impact of adolescent exposure to cannabinoids on resting state neural activity. In these experiments, 4 female adolescent rhesus macaques were repeatedly exposed to the CB1 full agonist AM2389. Resting state connectivity data was collected in a 3.0T fMRI before, during, and after 30 days following discontinuation of chronic drug treatment. This study used a multimodal imaging approach that generated structural, neurochemical, and functional data. In these studies, Dr. Kohut found regionally selective changes in the posterior singulate cortex during chronic AM2389 treatment, and magnetic resonance spectroscopy showed decreased n-acetylaspartate (NAA) levels in the medial orbital frontal cortex (mOFC) during acute and chronic treatment with AM2389 that recovered 30 days following discontinuation of chronic treatment. These changes in NAA levels were accompanied by functional changes between the mOFC and other brain regions. For instance, during acute treatments, there was decreased connectivity between the mOFC and the anterior cingulate; however, during chronic AM2389 treatment, there was increased connectivity between the mOFC and temporal brain regions (e.g., hippocampus) but decreased connectivity in regions associated with motoric behavior. Currently, these resting state studies are being extended to rhesus macaques self-administering various abused drugs within an fMRI. While this research is currently ongoing, early results have demonstrated that during cocaine self-administration, there is decreased activity in the putamen, premotor regions, and nucleus accumbens. These results are consistent with cocaine’s known pharmacological actions. Dr. Kohut plans to extend this research to elucidate neural activation patterns during self-administration of other abused drugs (e.g., nicotine, heroin). These developing studies and procedures are important as they can be used to evaluate neurobiological mechanisms of abused drugs and candidate medications. Overall, the research conducted and developed by Dr. Kohut provides an exciting new frontier in the fields of behavioral pharmacology and neuroscience. Results from these studies will provide key information on how drug exposure and drug-taking behavior interact and can be used to identify specific neural signatures associated to particular drugs of abuse. In turn, these unique neural signatures may be useful in identifying targets for the development of novel treatments to curtail addictive behaviors. Furthermore, studies conducted in awake, behaving monkeys will also deepen our understanding on drug-paired stimuli (e.g., lights that indicate responses will be reinforced with a drug infusion) impact drug-taking behavior, and may identify neurological indices of conditioned behavior (e.g., self-administration). Dr. Kohut has been attending CPDD annual meetings since 2006 and has been an associate member since 2011. Dr. Moura has been an associate member of CPDD since 2017 and currently serves on the Animals in Research Committee. By Fernando B. de Moura, Ph.D.- Animals in Research Committee Member
Opioid use disorders have risen to epidemic levels over the past two decades resulting in a dramatic increase in overdose death and addiction in the United States. Although opioid-based medications, such as methadone and buprenorphine, are available to help manage opioid use disorders, there are serious concerns over their safety and illicit diversion. These and other factors have contributed to suboptimal treatment outcomes and have prompted the search for safer and more effective non-opioid medication alternatives. Lorcaserin (BelviqR), a serotonin 2C receptor agonist and FDA-approved weight-loss medication, has been proposed as a candidate pharmacotherapy for abuse of stimulant drugs such as cocaine and amphetamines. However, the possible therapeutic effects of lorcaserin against other major drugs of abuse have remained largely a matter of conjecture. A recent study by Drs. Stephen Kohut and Jack Bergman from McLean Hospital and Harvard Medical School has shed new light on the potential therapeutic effectiveness of lorcaserin for the treatment of opioid use disorder. Using a well-validated nonhuman primate model of persistent heroin self-administration and employing a sophisticated behavioral economic analysis of heroin demand, these researchers established clear evidence that daily treatment with lorcaserin can significantly reduce the self-administration of heroin by decreasing its reinforcing strength as defined by the behavioral economic measure elasticity of demand. Lorcaserin did not, however, significantly alter the reinforcing strength of a palatable food reinforcer, which was used as a comparative control. These new preclinical findings are important because they show that lorcaserin can effectively decrease heroin demand and overall drug intake in a sustained, behaviorally selective manner. This profile of effects suggests that serotonin 2C receptor agonists such as lorcaserin may be prime candidates for the development of non-opioid alternatives for the management of opioid use disorder. Dr. Kohut has been an active member of CPDD since 2016 and has served on the Animals in Research Committee (2012-2016) and the Program Committee (2017-present). He is the recipient of the 2019 Joseph Cochin Young Investigator Award. Dr. Bergman has been an active member of CPDD since 1990. He has served in several capacities for the College and is currently Treasurer of CPDD and a member of the CPDD Board of Directors. Dr. Spealman has been a member of CPDD since 1992 and served as a Board of Director from 2010-2013. By Roger Spealman, Ph.D., Animals in Research Committee Member This research is published in the European Journal of Pharmacology 840: 28-32.
Cocaine use disorder (CUD) is a disorder with high rates of relapse. Despite these high rates of relapse, there are no current FDA-approved medications to prevent cocaine relapse. Two factors that have been shown to influence rates of relapse are the trait of impulsivity and an individual’s level of reactivity to cocaine-related cues. Understanding the underlying neural mechanisms of how both impulsivity and reactivity to cocaine cues influence cocaine relapse could potentially lead to better pharmacotherapeutic treatment options to enhance recovery from CUD. To investigate this question, Dr. Kathryn Cunningham and her colleagues at the University of Texas Medical Branch in Galveston have used various rodent animal models of both impulsivity and cue reactivity. Through their research, Dr. Cunningham’s team has shown that there is a common serotonergic pathway that underlies the ability of both impulsivity and cue-reactivity to serve as relapse vulnerability factors in CUD. In a recently published study in the Journal of Pharmacology and Experimental Therapeutics, Dr. Cunningham’s group looked specifically at the role of the 5-HT2A receptor in impulsive action and cocaine cue reactivity in rats. To exam this, the team first tested the effects of the FDA-approved, 5-HT2A receptor antagonist pimavanserin on the 1-choice serial reaction time task (1-CSRT) of impulsive action. They obtained baseline impulsive action on the 1-CSRT task in a second group of rats who then were tested for cocaine cue reactivity by allowing them to acquire stable levels of cocaine self-administration followed by forced abstinence for either 1 day or 30 days. Either one day or 30 days after cocaine abstinence, rats were pretreated with various doses of pimavanserin or saline prior to measurement of levels of drug-seeking responses. Dr. Cunningham’s group found that pimavanserin suppressed impulsive action, and that the baseline level of impulsive action predicted the levels of cocaine cue reactivity on day 30 of abstinence. They also found that baseline levels of impulsive action predicted the effectiveness of pimavanserin to decrease the level of cocaine cue reactivity after 30 days of abstinence. These data are particularly exciting as indicating a potential serotonergic drug, which already has FDA approval, could be useful as a treatment for relapse in CUD. Also, the utility of the drug to treat cocaine relapse may be increased if the individual’s level of impulsivity is also considered in the treatment plan. This research conducted in a preclinical, animal model is an important step in the process of medications development for CUD. Given the extensive time and expense required to move novel compounds toward clinical use, animal studies are helpful in optimizing clinical trials with regard to assessment methods and endpoints that serve as indicators of success or failure. Dr. Cunningham has a long history of participation and leadership in CPDD. She won a travel award to attend the 1988 CPDD meeting and became a CPDD member in 1993, and became a fellow in 2007. Dr. Cunningham has chaired the Publications and Nominating Committees, served on the Board of Directors, and was elected CPDD President and member of the Executive Committee. She was awarded the CPDD Mentorship Award (2012) and the Marian W. Fischman Memorial Award (2013). Dr. Stairs has been a member of CPDD since 2010. He currently serves as a member on the Animals in Research Committee. By Dustin J Stairs, Ph.D., Animals in Research Committee Member
Synthetic cannabinoids (i.e., “Spice”, “K2”) are emerging drugs of abuse that are associated with a host of adverse effects and visits to hospital emergency departments. In contrast to phytochemical cannabinoids derived from the cannabis plant (e.g., THC), less is known about the mechanisms responsible for their behavioral and physiological effects. Dr. William Fantegrossi, Associate Professor of Pharmacology and Toxicology at the University of Arkansas for Medical Sciences, uses rodent models to examine the neuropharmacological mechanisms contributing to their adverse effects. In a recent study (http://jpet.aspetjournals.org/content/early/2018/11/21/jpet.118.251157), Dr. Fantegrossi and his graduate student, Catheryn Wilson, examined the convulsant effects of two chemically dissimilar synthetic cannabinoids in mice. Seizures and convulsions are two of the most severe adverse effects associated with the use of synthetic cannabinoids in humans, but the mechanisms responsible for these effects are not fully understood, and potential treatments to reverse these effects have not been tested. Dr. Fantegrossi’s research team treated mice with the synthetic cannabinoid full agonists, JWH-018 and 5F-AB-PINACA, the phytochemical partial cannabinoid agonist, THC, and the non-cannabinoid chemical convulsant, pentylenetetrazol (PTZ). The two synthetic cannabinoids and PTZ reliably induced convulsant activity in a dose-dependent manner, whereas THC did not. Repeated administration of THC and JWH-018 produced tolerance to the convulsant activity of JWH-018 and 5F-AB-PINACA, but cross tolerance was not conferred to PTZ. Repeated administration of PTZ increased sensitivity to its convulsant effects, but did not alter the convulsant effects of synthetic cannabinoids. Importantly, the convulsant effects of synthetic cannabinoids were fully blocked by THC and the cannabinoid (CB1) receptor antagonist, rimonabant, but not by the prototypical anticonvulsant, diazepam. These findings indicate that that convulsant effects of two chemically dissimilar synthetic cannabinoids are both mediated by their strong agonist activity at CB1 receptors. The most clinically relevant finding of this study is that diazepam did not block the convulsant effects of either synthetic cannabinoid, even at a dose that completely blocked the convulsant effects of PTZ. Clinical reports of seizures and convulsions following the use of synthetic cannabinoids in humans usually report administration of diazepam and other benzodiazepines to medically manage these adverse effects. The findings obtained by Dr. Fantegrossi and his colleagues suggest that benzodiazepines may not be an effective treatment for these individuals, and suggest that acute administration of a CB1 antagonist like rimonabant might be a better therapeutic option. We know that cannabinoid-induced convulsions are consistent across species, given case reports of convulsions in domestic dogs and their owners following accidental or intentional ingestion of commercial Spice preparations. The animal model developed by Dr. Fantegrossi and his colleagues closely mimics the convulsant activity observed in human users. Cannabinoid-induced convulsions are typically witnessed by family or friends of the user at the time of use. Given the short duration of action of most synthetic cannabinoids, EEG readings (which are necessary to determine the presence of seizures) have typically returned to normal by the time the user arrives at the emergency department. Dr. Fantegrossi is currently conducting studies to measure the contributing role of seizures in eliciting cannabinoid-induced convulsant activity, which will represent a significant step forward in understanding the neurobiological mechanisms responsible for these effects. Dr. Fantegrossi has been a member of CPDD since 2001. He has previously served on the Electronics Committee, the Drug Evaluation Committee (2000-2006), and the Program Committee (2012-2014). He currently serves on the Committee for Abuse Liability Testing. Dr. Smith has been a member of CPDD since 1994. He currently serves on the Board of Directors and as Chair of the Animals in Research Committee. He previously served as Chair of the Awards Committee (2013-2017). By Mark Smith, Ph.D., Animals in Research Committee Chair
Epidemiological studies have shown repeatedly that one of the best prognosticators of adolescent or young adult drug use is whether an individual’s peers use drugs. The high concordance rates of drug use within peer groups can be explained by either selection theories or social learning theories. Selection theories propose that individuals choose or select their peers based on shared attitudes and behaviors regarding drug use. On the other hand, social learning theories propose that individuals learn drug use behaviors (or abstinent-related behaviors) by observing and imitating the behavior of their peers. Experimental studies addressing these theories have been limited because of a lack of preclinical models that allow laboratory animals to observe and mimic the drug use of others. To circumvent this problem, Dr. Mark Smith and his colleagues constructed custom-built, ‘social learning’, drug-administration chambers that enabled two animals to intravenously self-administer drugs at the same time and in the same chamber. Thus, these chambers allowed these animals to observe (and mimic) one another’s behavior. A wire screen prevented one rat from accessing the intravenous line and response lever of its partner. In order to determine the impact of observing a peer self-administer cocaine on the acquisition of cocaine use, Dr. Smith and colleagues assessed the behavior of three groups of rats. The first two groups were pair-housed and the third group was reared in isolation. For the first pair-housed group, one rat from each pair was trained to self-administer cocaine in isolation prior to reintroduction to its partner in the ‘social learning’ operant chamber where both rats had access to cocaine. For the second pair-housed group, neither had cocaine self-administration experience, and one rat from each pair did not have access to cocaine when placed in the ‘social learning’ operant chamber. The rats reared in isolation self-administered cocaine in standard (non-social-learning) test chambers. Dr. Smith and his colleagues found acquisition of cocaine self-administration was fastest among rats partnered with peers that had cocaine-self-administration experience, whereas rats partnered with peers that were not self-administering cocaine were the slowest to acquire cocaine self-administration. These findings demonstrate that observing a peer using cocaine has a profound impact on the development of drug use, whereas exposure to an abstinent peer is protective in delaying drug self-administration. As such, these findings provide evidence to support social learning theories underlying the development of drug use. The use of an animal model was an essential feature to this study that allowed the investigators to determine the cause-and-effect role that social contact with drug-using and abstinent peers have on the acquisition of drug use. These types of questions cannot be directly addressed in human populations, thus emphasizing the importance of animal models of human behavior in biomedical research. Dr. Smith has been a member of CPDD since 1994. He currently serves on the Board of Directors and as Chair of the Animals in Research Committee. He previously served as Chair of the Awards Committee (2013-2017). Dr. Cooper has been a member of CPDD since 2013. She is a member-elect of the Board of Directors and currently serves as a member on the Animals in Research Committee. She has previously served on the Program Committee (2014-2017) and Nominating Committee (2016-2017). By Ziva Cooper, Ph.D., Animals in Research Committee Member
Dr. Greg Collins is using procedures in laboratory animals to understand aspects related to designer psychostimulant abuse. The rise of designer recreational drugs has added further complication to our efforts to reduce drug addiction and abuse. These drugs are synthesized to mimic effects of existing recreational drugs, but often skirt detection and laws due to constantly changing, subtle alterations in their chemical structure. Of these new designer drugs, up to 20% are modified cathinones, often sold as ‘bath salts’ and marketed as alternatives to traditional psychostimulants like cocaine, methamphetamine, or MDMA. These drugs are commonly combined with other active ingredients in retail preparations; caffeine being among the most prominent co-constituents. Initially Dr. Collins expected that this adulteration simply represented a cost cutting measure by adding ‘filler’ to retail products. However, recent research from his preclinical laboratory indicates that the addition of caffeine to these products may impact their abuse liability and toxicity. The prototypical synthetic cathinones are MDPV and methylone, though over 140 related compounds have been identified in illicit use. Unlike traditional psychostimulants that act by inhibiting dopamine and serotonin reuptake back into neurons similarly, MDPV is more potent at inhibiting dopamine reuptake compared to serotonin. Based on our understanding of the critical role dopamine plays in the abuse liability of recreational drugs and the modulatory role serotonin plays, Dr. Collins hypothesized that MDPV might have even greater abuse liability than cocaine or methamphetamine. To explore this possibility, Dr. Collins uses intravenous self-administration procedures. He also uses these techniques to understand how the addition of caffeine to products containing synthetic cathinones impacts their abuse liability. In one procedure, laboratory rats press a lever a fixed number of times for each delivery of the drug or drug combination. In another, rats must make progressively more lever presses for each subsequent drug delivery. Using these techniques, Dr. Collins can assess the reinforcing potency and effectiveness of synthetic cathinones alone or combined with caffeine, and then compare these results to more traditional psychostimulants such as cocaine and methamphetamine. In his studies, Dr. Collins has found that synthetic cathinones maintain substantially higher amounts of behavior than cocaine or methamphetamine, indicating potentially greater abuse liability. Using dose-addition analysis, he also found that the addition of caffeine further enhances the effectiveness of synthetic cathinones to maintain behavior, and also increases their potency to do so. This suggests that caffeine may be an important constituent in these products that may further increase their misuse. This is consistent with clinical reports that cathinone products can engender prolonged bouts of use that exceed the patterns of use of more traditional psychostimulants. Dr. Collins is beginning to investigate other aspects of this interaction, particularly the potential that combinations of synthetic cathinones and caffeine are more toxic than either drug alone. To this end, he has begun assessing cardiopulmonary effects of these drugs and their combination. If these combinations are both more liable to abuse and produce greater toxicity, it would focus efforts on increasing awareness about these potential problems among psychostimulant users. Dr. Collins’ preclinical research is important in several ways. It is illuminating an often under-appreciated aspect of substance abuse; that other constituents in a product can interact with the primary ingredient to produce unexpected and often detrimental effects. It also represents an attempt to better model patterns of human substance use in animal models, by examining pharmacological interactions that occur in polysubstance abuse, the most common pattern seen in humans. Dr. Collins has regularly attended the CPDD annual meeting since 2004 and has been a regular member since 2011. He is active as a member of the Programming Committee. Dr. Ginsburg has been a member of CPDD since 2006 and currently serves on the Animals in Research Committee. By Brett Ginsburg, Ph.D., Animals in Research Committee Chair
Tobacco use is the greatest preventable cause of death, cancer, and illness in the United States. Some smoking cessation interventions are effective, but the majority of individuals who stop smoking relapse within a year. A better understanding of the mechanisms underlying vulnerability to relapse can help pave the way for more effective treatments. Mark Namba and Dr. Cassie Gipson-Reichardt in the Neurobiology and Behavior in Addiction Lab at Arizona State University use an animal model of cue-induced relapse to study neurobiological substrates underlying relapse vulnerability. Under this model, rats are trained to self-administer nicotine, the primary active alkaloid in tobacco. Then, rats are placed into extinction training for several days, in which self-administration responses no longer produce nicotine or nicotine-paired cues. Following extinction, cue-induced nicotine relapse is tested by allowing the rats to respond in the presence of cues that had previously been paired with nicotine delivery. In an ongoing study, Namba and Dr. Gipson-Reichardt are using this model to examine whether drug-induced neuroinflammation may underlie vulnerability to nicotine relapse. Chronic nicotine self-administration is associated with enduring alterations in glutamatergic plasticity within the nucleus accumbens core, including dysregulation of glial glutamate transport. The nuclear factor kappa B (NF-κB) pathway mediates drug-induced neuroinflammation and is a key regulator of synaptic plasticity. Notably, little is known about NF-κB’s role in cue-induced nicotine relapse. To address this limitation, Namba and Dr. Gipson-Reichardt are using a herpes virus gene transfer strategy to express several forms of IκB kinase (IKK) to bi-directionally modulate activation of downstream NF-κB. Additionally, they and others have demonstrated that N-acetylcysteine, an antioxidant and glutamatergic agent, inhibits cue-induced nicotine relapse and are currently examining whether NF-κB signaling underlies its therapeutic efficacy. Preliminary results suggest that NF-κB signaling mediates cue-induced nicotine relapse. Specifically, inhibition of NF-κB inhibits cue-induced reinstatement, whereas activation of NF-κB drives reinstatement and blocks the attenuating effect of N-acetylcysteine on reinstatement. Overall, these findings are significant because they highlight the potential role for neuroimmune signaling as a modulatory mechanism that regulates glutamatergic synaptic plasticity and subsequent drug-seeking behavior, thus revealing a new avenue for the development of novel pharmacotherapeutics aimed at reducing the risk of nicotine relapse. Dr. Gipson-Reichardt has been a member of CPDD since 2013; she currently serves on the Program Committee. Dr. Holtyn has been a member of CPDD since 2013; she currently serves on the Animals in Research Committee. By August Holtyn, Ph.D., Animals in Research Committee Chair *This work was supported by the National Institutes of Health Grant DA036569 and -S1.
Although cocaine use is a leading cause of overdose deaths in the US, second only to opioids, there are currently no FDA-approved medications for its treatment. Exercise shows promise as a treatment option given its potential to reduce drug craving and vulnerability to relapse, but studies conducted in clinical populations have yield variable findings. Dr. Wendy Lynch and her colleagues at the University of Virginia use an animal model of relapse to examine behavioral and pharmacological treatments that might reduce relapse vulnerability. Dr. Lynch trains laboratory rats to self-administer cocaine by pressing a response lever. Later, cocaine is removed and an exercise wheel is introduced during either early or late abstinence. Relapse vulnerability is then tested by allowing the rats to respond on the lever in the presence of cues that had previously been predictive of cocaine delivery. Using this model, Dr. Lynch and her colleagues recently reported that exercise introduced early, but not late, during abstinence is critical for its efficacy to reduce relapse vulnerability. They then determined the mechanism by which exercise introduced early during abstinence may reduce relapse to cocaine use. Dr. Lynch and her colleagues found that the protective effects of exercise were associated with metabotropic glutamate receptor 5 expression (mGluR5) in the prefrontal cortex. Glutamate signaling in the prefrontal cortex is known to be involved in the development of compulsive drug use, and this finding suggests that exercise may be producing its protective effects by altering signaling in this area. To examine this possibility, they injected a mGluR5 receptor agonist directly into the prefrontal cortex during early abstinence and then tested the rats in their model of relapse. As they predicted, stimulation of mGluR5 receptors in the prefrontal cortex simulated the protective effect of early-initiated exercise and reduced relapse vulnerability. These results are the first to show that exercise initiated during early abstinence functions as an effective treatment intervention for cocaine addiction by normalizing dysregulated mGluR5 signaling in the prefrontal cortex. The use of an animal model of relapse allowed Dr. Lynch to determine the causal mechanisms by which exercise was producing its protective effects on relapse vulnerability. These findings are important for the development of treatment interventions in human populations by showing that relapse can be prevented through exercise and/or medications that upregulate glutamatergic signaling in the prefrontal cortex during early abstinence. Dr. Lynch has been a member of CPDD since 1997; she has previously served on the Program Committee (2011-2013) and the Animals in Research Committee (2009-2012). Dr. Smith has been a member of CPDD since 1994. He currently serves on the Board of Directors and as Chair of the Animals in Research Committee. He previously served as Chair of the Awards Committee (2013-2017). By Mark Smith, Ph.D., Animals in Research Committee Chair
Opioid analgesics remain the primary treatment for moderate to severe pain, but often cause behavioral impairment that constrains their therapeutic utility. Preclinical antinociception assays in animals have been indispensable in the development of analgesic medications, but have not typically provided concurrent assessment of behaviorally disruptive effects. To address this shortcoming, Sarah Withey and colleagues Carol Paronis and Jack Bergman of the Preclinical Pharmacology Laboratory of McLean Hospital, Harvard Medical School modified a previously validated warm-water tail withdrawal assay to measure tail withdrawal latencies (antinociception) concurrently with disruption of food-reinforced operant responding (behavioral impairment). A total of six different opioids were tested, and each resulted in dose-dependent antinociception and varying degrees of behavioral impairment. The researchers used a preclinical therapeutic ratio as a measure of a drug’s behavioral selectivity, which was defined as the ED50 for behavioral impairment divided by the ED50 for antinociception. The preclinical therapeutic ratio differed widely among the drugs. Nalbuphine had the highest therapeutic ratio (4.88) indicating significant antinociception at doses that did not cause behavioral impairment, whereas butorphanol had the lowest therapeutic ratio (0.17) indicating significant behavioral impairment at doses that were not sufficient to produce antinociception. Other opioids, including oxycodone, heroin, buprenorphine and methadone, had therapeutic ratios approaching 1.0, indicating that the antinociceptive and behaviorally impairing effects were produced by similar doses. These results are important because they demonstrate the utility of this animal model to concurrently measure antinociception and behavioral impairment, which may provide a useful technique for predicting the selectivity with which novel analgesics exert their therapeutic effects. Dr. Withey is new to the CPDD family; Dr. Bergman has been a member of CPDD since 1993 and currently serves as the Treasurer of the organization; Dr. Spealman has been a member of CPDD since 1992 and served as a Board of Director from 2010-2013. By Roger Spealman, Ph.D., Animals in Research Committee Member This research is published in The Journal of Pain.
Stress is an important predictor of substance use and misuse. For example, early-life or chronic stress can contribute to the development of disordered patterns of substance use. Similarly, stressful events may precipitate relapse and continued substance use among recovering individuals. However, not everyone reacts to the effects of stress equally. Animal models can help provide insight into the behavioral, physiological, and neural correlates underlying differential responses to environmental events, such as stress. New research by Dr. Michael Nader and colleagues at Wake Forest School of Medicine uses a nonhuman primate model of cocaine use to show that social rank is an important factor related to the behavioral and biological response to social stress. This study is a part of Dr. Nader’s broader research program evaluating the neurobiological and behavioral consequences of social status on measures relevant to cocaine and other substance use disorders. The first experiment in this study began by examining cocaine self-administration in socially housed, male cynomolgus macaques using a food-drug choice procedure. This food-drug choice procedure provides a translationally relevant measure of substance use by modeling the decision to use drugs in the presence of a competing non-drug reinforcer (i.e., food). Subjects were also divided into a dominant or subordinate group based on social behaviors observed in the home cage. Dominant and subordinate monkeys were tested under conditions such that similar rates of self-administration were observed under these “baseline” conditions. Following baseline testing, subjects were exposed to social stress using a resident/intruder procedure. In this procedure, subjects were placed into another social group as an “intruder” for 30 minutes (note that the resident monkeys could not physically contact the intruder). The effects of this social stress on cocaine self-administration were then determined using the same food-drug choice paradigm. A majority of subjects were affected by the social stress manipulation, however the direction of this effect depended on the monkey’s social rank. Specifically, being an intruder in another social group produced increases in sensitivity to the reinforcing effects of cocaine in subordinate monkeys, but produced decreases in sensitivity to the reinforcing effects of cocaine in dominant monkeys. The former effects resemble the consequences of stress, while the latter outcome is what would be hypothesized to occur following an enriching event. Importantly, the same environmental manipulation had different consequences depending on the social rank of the monkey. A second experiment in this study attempted to reveal what neurobiological mechanisms mediated this social rank-stress relationship. To this end, PET imaging was used to study brain glucose metabolism in dominant and subordinate monkeys at baseline and following intruder stress. Clear differences between the dominant and subordinate monkeys were observed under both conditions. At baseline, dominant monkeys showed greater activity in regions associated with visual processing, attentional control, and vigilance, whereas subordinate monkey showed greater activity in regions associated with emotional processing, fear, and anxiety. Social stress increased activity along the HPA-axis in both groups with enhanced activity in the posterior cingulate cortex (a region associated with risk to cocaine relapse) in subordinate monkeys. Dr. Nader and colleagues’ study highlights the importance of preclinical animal models for identifying individual phenotypes conferring resilience or susceptibility to environmental events relevant to substance use. Although food-drug choice behavior was similar between dominant and subordinate monkeys under baseline conditions, acute social stress produced marked differences in responding based on a subject’s social rank. Animal models, such as these, can help detect clinically relevant factors underlying different patterns of behavior observed in the human clinical condition. Such an approach can also reveal behavioral, physiological, and neural markers to promote individualized approaches in treating substance use disorder. Michael Nader has been a member of CPDD since 1988 and served on the Board of Directors from 2006-2010, the Drug Testing and Evaluation Liaison Committee (2000-2003; 2007-2010), being Chair of that committee from 2007-2009, and is a past member of the Nomination Committee (2010). Justin Strickland has been a member of CPDD since 2014 and currently serves on the Animals in Research and Travel Award committees. By Justin C. Strickland, M.S., Animals in Research Committee Member The study was supported by the National Institute on Drug Abuse.