1
|
Herman RJ, Schmidt HD. Targeting GLP-1 receptors to reduce nicotine use disorder: Preclinical and clinical evidence. Physiol Behav 2024; 281:114565. [PMID: 38663460 PMCID: PMC11128349 DOI: 10.1016/j.physbeh.2024.114565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Nicotine use disorder (NUD) remains a leading cause of preventable death in the U.S. Unfortunately, current FDA-approved pharmacotherapies for smoking cessation have limited efficacy and are associated with high rates of relapse. One major barrier to long-term smoking abstinence is body weight gain during withdrawal. Nicotine withdrawal-induced body weight gain can also lead to development of chronic disease states like obesity and type II diabetes mellitus. Therefore, it is critical to identify novel pharmacotherapies for NUD that decrease relapse and nicotine withdrawal symptoms including body weight gain. Recent studies demonstrate that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate voluntary nicotine taking and seeking and prevent withdrawal-induced hyperphagia and body weight gain. Emerging evidence also suggests that GLP-1R agonists improve cognitive deficits, as well as depressive- and anxiety-like behaviors, which contribute to smoking relapse during withdrawal. While further studies are necessary to fully characterize the effects of GLP-1R agonists on NUD and understand the mechanisms by which GLP-1R agonists decrease nicotine withdrawal-mediated behaviors, the current literature supports GLP-1R-based approaches to treating NUD.
Collapse
Affiliation(s)
- Rae J Herman
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
2
|
Tripathi PN, Lodhi A, Rai SN, Nandi NK, Dumoga S, Yadav P, Tiwari AK, Singh SK, El-Shorbagi ANA, Chaudhary S. Review of Pharmacotherapeutic Targets in Alzheimer's Disease and Its Management Using Traditional Medicinal Plants. Degener Neurol Neuromuscul Dis 2024; 14:47-74. [PMID: 38784601 PMCID: PMC11114142 DOI: 10.2147/dnnd.s452009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. While there is currently no cure for AD, several pharmacotherapeutic targets and management strategies have been explored. Additionally, traditional medicinal plants have gained attention for their potential role in AD management. Pharmacotherapeutic targets in AD include amyloid-beta (Aβ) aggregation, tau protein hyperphosphorylation, neuroinflammation, oxidative stress, and cholinergic dysfunction. Traditional medicinal plants, such as Ginkgo biloba, Huperzia serrata, Curcuma longa (turmeric), and Panax ginseng, have demonstrated the ability to modulate these targets through their bioactive compounds. Ginkgo biloba, for instance, contains flavonoids and terpenoids that exhibit neuroprotective effects by reducing Aβ deposition and enhancing cerebral blood flow. Huperzia serrata, a natural source of huperzine A, has acetylcholinesterase-inhibiting properties, thus improving cholinergic function. Curcuma longa, enriched with curcumin, exhibits anti-inflammatory and antioxidant effects, potentially mitigating neuroinflammation and oxidative stress. Panax ginseng's ginsenosides have shown neuroprotective and anti-amyloidogenic properties. The investigation of traditional medicinal plants as a complementary approach to AD management offers several advantages, including a lower risk of adverse effects and potential multi-target interactions. Furthermore, the cultural knowledge and utilization of these plants provide a rich source of information for the development of new therapies. However, further research is necessary to elucidate the precise mechanisms of action, standardize preparations, and assess the safety and efficacy of these natural remedies. Integrating traditional medicinal-plant-based therapies with modern pharmacotherapies may hold the key to a more comprehensive and effective approach to AD treatment. This review aims to explore the pharmacotherapeutic targets in AD and assess the potential of traditional medicinal plants in its management.
Collapse
Affiliation(s)
- Prabhash Nath Tripathi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Ankit Lodhi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Sachchida Nand Rai
- Center of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Nilay Kumar Nandi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Shweta Dumoga
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Pooja Yadav
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Amit Kumar Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Santosh Kumar Singh
- Center of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Abdel-Nasser A El-Shorbagi
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Sachin Chaudhary
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
3
|
Shreya S, Annamalai M, Jirge VL, Sethi S. Utility of salivary biomarkers for diagnosis and monitoring the prognosis of nicotine addiction - A systematic review. J Oral Biol Craniofac Res 2023; 13:740-750. [PMID: 38028231 PMCID: PMC10630637 DOI: 10.1016/j.jobcr.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 10/08/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Tobacco and smoke associated with tobacco comprises of a mixture of more than 9500 chemical compounds, most of which have been identified as harmful. Two of the most potent carcinogens found in cigarette smoke are N'-nitrosonornicotine (NNN) and polycyclic aromatic hydrocarbons (PAHs). The most commonly used method to detect and monitor nicotine addiction is via serum cotinine levels. Though considered the gold standard, there is a decline in preventive screening and diagnostic testing due to the fear of pain from invasive testing. Data sources and study selection A structured literature search was performed using the search engines PubMed and Google scholar following the PRISMA guidelines for systematic reviews. The titles and abstracts were retrieved and analysed, followed by full-text relevant data extraction in addition to a risk-of-bias analysis. Data extraction and synthesis A total of 37 studies were included in the systematic review. Salivary cotinine levels were compared between smokers and non-smokers, cigarette smokers and water pipe smokers, water pipe smokers and non-smokers. Lactate dehydrogenase salivary levels were compared between smokers and non-smokers, and salivary thiocyanate were compared between smokers and non-smokers. Conclusions Identifying biomarkers with high performance in terms of sensitivity and specificity will contribute to accelerating future research in this domain.
Collapse
Affiliation(s)
- S. Shreya
- A B Shetty Memorial Institute of Dental Sciences, Mangalore, Karnataka, India
| | - Manoj Annamalai
- KAHER’S KLE V.K. Institute of Dental Sciences, Belagavi, Karnataka, 590010, India
| | - Vasanti Lagali Jirge
- KAHER’S KLE V.K. Institute of Dental Sciences, Belagavi, Karnataka, 590010, India
| | - Sneha Sethi
- Adelaide Dental School, University of Adelaide, Adelaide, 5000, Australia
| |
Collapse
|
4
|
Hosseini SH, Khabbazhosseini ZS, Khatibi S, Yahosseini A, Borhaninejad N, Beheshti F, Kakhki S. Folic acid supplementation improved nicotine withdrawal-induced of memory loss via affecting oxidative status, inflammatory response, cholinergic activity, BDNF and amyloid-B in adolescent male rat. Neurosci Lett 2023; 815:137489. [PMID: 37741611 DOI: 10.1016/j.neulet.2023.137489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND The present study aimed to assess whether folic acid (FA) have potential to prevent memory impairment caused by nicotine (Nico) withdrawal in adolescent male rats. METHODS AND MATERIALS The experiments were divided into 7 groups: 1) vehicle, 2) Nico (Nico 2 mg/kg injection from 21 to 42 days of ages), 3-5) Nico FA5/10/15 mg/kg (received Nico from 21 to 42 days of ages and received FA at three doses 5, 10 and 15 mg/kg 43-63 days of ages), and 6) received normal saline from 21 to 42 days of age after that received FA 15 mg/kg by oral gavage from 43 to 63 days of age. At 64-69 days of ages, behavioral tests related to memory including Morris Water Maze (MWM) and Object Recognition Test (ORT) were performed and related biochemical analysis including the hippocampal levels of oxidative stress markers, inflammatory indices, brain-derived neurotrophic factor (BDNF), nitrite, amyloid-B and acetylcholinesterase [1] were measured. RESULTS Results showed that nicotine exposure in adolescence followed by withdrawal dramatically impaired learning and memory performance along with affecting a variety of biochemical markers in the hippocampal tissues. In addition, it was observed that administration of FA significantly ameliorated Nico withdrawal-induced adverse effects through restoration of the mentioned biochemical disturbances. CONCLUSION The present study and other relevant researches demonstrated that FA as a well-known, inexpensive, and safe supplement has strong potential to either prevent or ameliorate the detrimental effect of Nico withdrawal. However, further investigation is required to be more elucidated the precise mechanisms underlying memory impairment-induced by Nico withdrawal.
Collapse
Affiliation(s)
- Seyed Hossein Hosseini
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | | | - Seyedamirsadra Khatibi
- Student Research Committee, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirreza Yahosseini
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Nafiseh Borhaninejad
- Student Research Committee, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farimah Beheshti
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Departments of Physiology, School of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| | - Samaneh Kakhki
- Department of Clinical Biochemistry, School of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| |
Collapse
|
5
|
Herman RJ, Hayes MR, Audrain-McGovern J, Ashare RL, Schmidt HD. Liraglutide attenuates nicotine self-administration as well as nicotine seeking and hyperphagia during withdrawal in male and female rats. Psychopharmacology (Berl) 2023; 240:1373-1386. [PMID: 37129617 PMCID: PMC11088902 DOI: 10.1007/s00213-023-06376-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
RATIONALE Nicotine cessation is associated with increased consumption of highly palatable foods and body weight gain in most smokers. Concerns about body weight gain are a major barrier to maintaining long-term smoking abstinence, and current treatments for nicotine use disorder (NUD) delay, but do not prevent, body weight gain during abstinence. Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce food intake and are FDA-approved for treating obesity. However, the effects of GLP-1R agonist monotherapy on nicotine seeking and withdrawal-induced hyperphagia are unknown. OBJECTIVES We screened the efficacy of the long-lasting GLP-1R agonist liraglutide to reduce nicotine-mediated behaviors including voluntary nicotine taking, as well as nicotine seeking and hyperphagia during withdrawal. METHODS Male and female rats self-administered intravenous nicotine (0.03 mg/kg/inf) for ~21 days. Daily liraglutide administration (25 μg/kg, i.p.) started on the last self-administration day and continued throughout the extinction and reinstatement phases of the experiment. Once nicotine taking was extinguished, the reinstatement of nicotine-seeking behavior was assessed after an acute priming injection of nicotine (0.2 mg/kg, s.c.) and re-exposure to conditioned light cues. Using a novel model of nicotine withdrawal-induced hyperphagia, intake of a high fat diet (HFD) was measured during home cage abstinence in male and female rats with a history of nicotine self-administration. RESULTS Liraglutide attenuated nicotine self-administration and reinstatement in male and female rats. Repeated liraglutide attenuated withdrawal-induced hyperphagia and body weight gain in male and female rats at a dose that was not associated with malaise-like effects. CONCLUSIONS These findings support further studies investigating the translational potential of GLP-1R agonists to treat NUD.
Collapse
Affiliation(s)
- R J Herman
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, TRL Room 2215, Philadelphia, PA, 19104, USA
| | - J Audrain-McGovern
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, TRL Room 2215, Philadelphia, PA, 19104, USA
| | - R L Ashare
- Department of Psychology, College of Arts and Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - H D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, TRL Room 2215, Philadelphia, PA, 19104, USA.
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, 125 South 31st Street, TRL Room 2215, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Perkins KA. The 2022 Ferno Award Address: CrEATE, an Efficient Crossover Evaluation of Addiction Treatment Efficacy. Nicotine Tob Res 2023; 25:77-85. [PMID: 35671343 PMCID: PMC9717395 DOI: 10.1093/ntr/ntac139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/05/2022] [Accepted: 06/05/2022] [Indexed: 01/03/2023]
Abstract
Dozens of drugs have been evaluated in recent decades for initial evidence of efficacy to aid smoking cessation (i.e. "early Phase 2" testing, according to U.S. FDA terminology), with the vast majority failing to show efficacy. Even small randomized clinical trials (RCTs), the most common early Phase 2 tests, are costly undertakings, made more unappealing by their high likelihood of failure. At the same time, another early Phase 2 approach, acute tests of drug effects on surrogate endpoints such as withdrawal or craving severity, are more practical but have little predictive clinical validity. Described here is an innovative procedure that optimally combines the validity of clinical trials with the practical advantages of surrogate endpoint studies to more efficiently determine whether or not a novel drug warrants continued clinical development. This CrEATE procedure, or Crossover Evaluation of Addiction Treatment Efficacy, does so by assessing short-term quit success in smokers highly motivated to quit when briefly treated with active drug versus placebo in a crossover design, so that quit efficacy from both conditions is compared within participants. The program to develop and evaluate CrEATE demonstrates its sensitivity to efficacy from all three FDA-approved first-line cessation medications (NRT, varenicline, bupropion), tested here as model drugs, as well as specificity in identifying lack of efficacy with a drug known to be ineffective for cessation (modafinil). CrEATE has subsequently been used to evaluate a few novel interventions, concluding they lack efficacy in increasing quit success. Future directions for the potential utility of CrEATE are provided. Implications: The ability of CrEATE to reach a Go/No Go decision more quickly and with far less cost lowers the risk of failure, meaning widespread use of the procedure should encourage the evaluation of more novel candidate drugs. With its greater efficiency, failed tests, unfortunately the most likely outcome in early Phase 2 studies, will cause less waste of resources. At the same time, CrEATE tests that indicate a novel treatment has efficacy will justify the substantial time and expense of moving forward to evaluate the drug in late Phase 2 RCTs.
Collapse
Affiliation(s)
- Kenneth A Perkins
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania PA 15213, USA
| |
Collapse
|
7
|
Williams AV, Flowers J, Coates KS, Ingram A, Hehn AT, Dupuis M, Wimmer ME, Venniro M, Bangasser DA. Early resource scarcity alters motivation for natural rewards in a sex- and reinforcer-dependent manner. Psychopharmacology (Berl) 2022; 239:3929-3937. [PMID: 36301314 PMCID: PMC9817039 DOI: 10.1007/s00213-022-06264-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/13/2022] [Indexed: 01/11/2023]
Abstract
RATIONALE Early life adversity impacts reward-related behaviors, including reward seeking for drugs of abuse. However, the effects of early stress on natural rewards, such as food and social rewards, which have strong implications for symptoms of psychiatric conditions such as major depressive disorder (MDD), are understudied. To fill this gap, we used the limited bedding and nesting (LBN) procedure to assess the impact of early resource scarcity on motivational drive for both food and social rewards in rats. METHODS Male and female Long Evans rats were reared in either an LBN environment, with limited nesting materials and no enrichment, from their postnatal day 2-9 or control environment with ample nesting materials and enrichment. As adults, they were tested for reward-seeking behavior on progressive ratio operant tasks: food reward (sucrose) or social reward (access to a same-sex/age conspecific). RESULTS We observed sex differences in the impact of LBN on motivation for natural rewards. In males, LBN increased motivation for both a sucrose and social reward. In females, LBN reduced motivation for sucrose but had no effect on social reward. CONCLUSIONS These results suggest that the effects of LBN on motivation for natural rewards are both sex- and reinforcer-dependent, with males and females showing differential motivation for food and social rewards following early scarcity. Our previous data revealed an LBN-driven reduction in motivation for morphine in males and no effect in females, highlighting the reinforcer-dependent impact of early resource scarcity on motivated behavior more widely.
Collapse
Affiliation(s)
- Alexia V Williams
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - James Flowers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Kennedy S Coates
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Atiba Ingram
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Alexandra T Hehn
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Molly Dupuis
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA
| | - Marco Venniro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
- Neuroscience Institute, Georgia State University, GA, 30303-5030, Atlanta, USA.
| |
Collapse
|
8
|
Maurer JJ, Wimmer ME, Turner CA, Herman RJ, Zhang Y, Ragnini K, Ferrante J, Kimmey BA, Crist RC, Christopher Pierce R, Schmidt HD. Paternal nicotine taking elicits heritable sex-specific phenotypes that are mediated by hippocampal Satb2. Mol Psychiatry 2022; 27:3864-3874. [PMID: 35595980 PMCID: PMC9675874 DOI: 10.1038/s41380-022-01622-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023]
Abstract
Nicotine intake, whether through tobacco smoking or e-cigarettes, remains a global health concern. An emerging preclinical literature indicates that parental nicotine exposure produces behavioral, physiological, and molecular changes in subsequent generations. However, the heritable effects of voluntary parental nicotine taking are unknown. Here, we show increased acquisition of nicotine taking in male and female offspring of sires that self-administered nicotine. In contrast, self-administration of sucrose and cocaine were unaltered in male and female offspring suggesting that the intergenerational effects of paternal nicotine taking may be reinforcer specific. Further characterization revealed memory deficits and increased anxiety-like behaviors in drug-naive male, but not female, offspring of nicotine-experienced sires. Using an unbiased, genome-wide approach, we discovered that these phenotypes were associated with decreased expression of Satb2, a transcription factor known to play important roles in synaptic plasticity and memory formation, in the hippocampus of nicotine-sired male offspring. This effect was sex-specific as no changes in Satb2 expression were found in nicotine-sired female offspring. Finally, increasing Satb2 levels in the hippocampus prevented the escalation of nicotine intake and rescued the memory deficits associated with paternal nicotine taking in male offspring. Collectively, these findings indicate that paternal nicotine taking produces heritable sex-specific molecular changes that promote addiction-like phenotypes and memory impairments in male offspring.
Collapse
Affiliation(s)
- John J Maurer
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mathieu E Wimmer
- Department of Psychology, College of Liberal Arts, Temple University, Philadelphia, PA, 19122, USA
| | - Christopher A Turner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rae J Herman
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yafang Zhang
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kael Ragnini
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julia Ferrante
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Blake A Kimmey
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Richard C Crist
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Chang YP, Ma X, Shao H, Zhao YM. Total Syntheses of Galanthamine and Lycoramine via a Palladium-Catalyzed Cascade Cyclization and Late-Stage Reorganization of the Cyclized Skeleton. Org Lett 2021; 23:9659-9663. [PMID: 34874174 DOI: 10.1021/acs.orglett.1c03943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Herein, we report the highly efficient total syntheses of galanthamine and lycoramine from a common tetracyclic intermediate. This concise synthetic route features a two-phase strategy, which includes the early-stage rapid construction of a tetracyclic skeleton followed by the late-stage selective reorganization of the tetracyclic skeleton. Key to the success of this strategy are a palladium-catalyzed carbonylative cascade annulation, a DDQ-mediated intramolecular regioselective oxidative lactamization, as well as a BF3·Et2O-promoted reorganization of the bridged tetracyclic skeleton.
Collapse
Affiliation(s)
- Ya-Ping Chang
- Key Laboratory of Applied Surface and Colloid Chemistry & School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Ave, Xi'an, 710119, China
| | - Xia Ma
- Key Laboratory of Applied Surface and Colloid Chemistry & School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Ave, Xi'an, 710119, China
| | - Hui Shao
- Key Laboratory of Applied Surface and Colloid Chemistry & School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Ave, Xi'an, 710119, China
| | - Yu-Ming Zhao
- Key Laboratory of Applied Surface and Colloid Chemistry & School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Ave, Xi'an, 710119, China.,CAS Key Lab of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Ling Ling Road, Shanghai 200032, China
| |
Collapse
|
10
|
Curcumin Potentiates α7 Nicotinic Acetylcholine Receptors and Alleviates Autistic-Like Social Deficits and Brain Oxidative Stress Status in Mice. Int J Mol Sci 2021; 22:ijms22147251. [PMID: 34298871 PMCID: PMC8303708 DOI: 10.3390/ijms22147251] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022] Open
Abstract
Autistic spectrum disorder (ASD) refers to a group of neurodevelopmental disorders characterized by impaired social interaction and cognitive deficit, restricted repetitive behaviors, altered immune responses, and imbalanced oxidative stress status. In recent years, there has been a growing interest in studying the role of nicotinic acetylcholine receptors (nAChRs), specifically α7-nAChRs, in the CNS. Influence of agonists for α7-nAChRs on the cognitive behavior, learning, and memory formation has been demonstrated in neuro-pathological condition such as ASD and attention-deficit hyperactivity disorder (ADHD). Curcumin (CUR), the active compound of the spice turmeric, has been shown to act as a positive allosteric modulator of α7-nAChRs. Here we hypothesize that CUR, acting through α7-nAChRs, influences the neuropathology of ASD. In patch clamp studies, fast inward currents activated by choline, a selective agonist of α7-nAChRs, were significantly potentiated by CUR. Moreover, choline induced enhancement of spontaneous inhibitory postsynaptic currents was markedly increased in the presence of CUR. Furthermore, CUR (25, 50, and 100 mg/kg, i.p.) ameliorated dose-dependent social deficits without affecting locomotor activity or anxiety-like behaviors of tested male Black and Tan BRachyury (BTBR) mice. In addition, CUR (50 and 100 mg/kg, i.p.) mitigated oxidative stress status by restoring the decreased levels of superoxide dismutase (SOD) and catalase (CAT) in the hippocampus and the cerebellum of treated mice. Collectively, the observed results indicate that CUR potentiates α7-nAChRs in native central nervous system neurons, mitigates disturbed oxidative stress, and alleviates ASD-like features in BTBR mice used as an idiopathic rodent model of ASD, and may represent a promising novel pharmacological strategy for ASD treatment.
Collapse
|
11
|
Moerke MJ, McMahon LR, Wilkerson JL. More than Smoke and Patches: The Quest for Pharmacotherapies to Treat Tobacco Use Disorder. Pharmacol Rev 2020; 72:527-557. [PMID: 32205338 DOI: 10.1124/pr.119.018028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tobacco use is a persistent public health issue. It kills up to half its users and is the cause of nearly 90% of all lung cancers. The main psychoactive component of tobacco is nicotine, primarily responsible for its abuse-related effects. Accordingly, most pharmacotherapies for smoking cessation target nicotinic acetylcholine receptors (nAChRs), nicotine's major site of action in the brain. The goal of the current review is twofold: first, to provide a brief overview of the most commonly used behavioral procedures for evaluating smoking cessation pharmacotherapies and an introduction to pharmacokinetic and pharmacodynamic properties of nicotine important for consideration in the development of new pharmacotherapies; and second, to discuss current and potential future pharmacological interventions aimed at decreasing tobacco use. Attention will focus on the potential for allosteric modulators of nAChRs to offer an improvement over currently approved pharmacotherapies. Additionally, given increasing public concern for the potential health consequences of using electronic nicotine delivery systems, which allow users to inhale aerosolized solutions as an alternative to smoking tobacco, an effort will be made throughout this review to address the implications of this relatively new form of nicotine delivery, specifically as it relates to smoking cessation. SIGNIFICANCE STATEMENT: Despite decades of research that have vastly improved our understanding of nicotine and its effects on the body, only a handful of pharmacotherapies have been successfully developed for use in smoking cessation. Thus, investigation of alternative pharmacological strategies for treating tobacco use disorder remains active; allosteric modulators of nicotinic acetylcholine receptors represent one class of compounds currently under development for this purpose.
Collapse
Affiliation(s)
- M J Moerke
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| | - L R McMahon
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| | - J L Wilkerson
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| |
Collapse
|
12
|
Smith LC, George O. Advances in smoking cessation pharmacotherapy: Non-nicotinic approaches in animal models. Neuropharmacology 2020; 178:108225. [PMID: 32758566 DOI: 10.1016/j.neuropharm.2020.108225] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/22/2022]
Abstract
The landscape of worldwide tobacco use is changing, with a decrease in traditional smoking and an exponential rise in electronic cigarette use. No new nicotine cessation pharmacotherapies have come to market in the last 10 years. The current therapies that have been approved by the United States Food and Drug Administration for nicotine cessation include nicotine replacement therapy, varenicline, a nicotinic acetylcholine receptor partial agonist, and the atypical antidepressant bupropion. Nicotine replacement therapy and varenicline both act on nicotinic acetylcholine receptors. Bupropion inhibits the dopamine transporter, the norepinephrine transporter, and the nicotinic acetylcholine receptors to inhibit smoking behavior. Notwithstanding these treatments, rates of successful nicotine cessation in clinical trials remain low. Recent pharmacological approaches to improve nicotine cessation rates in animal models have turned their focus away from activating nicotinic acetylcholine receptors. The present review focuses on such pharmacological approaches, including nicotine vaccines, anti-nicotine antibodies, nicotine-degrading enzymes, cannabinoids, and metformin. Both immunopharmacological and enzymatic approaches rely on restricting and degrading nicotine within the periphery, thus preventing psychoactive effects of nicotine on the central nervous system. In contrast, pharmacologic inhibition of the enzymes which degrade nicotine could affect smoking behavior. Cannabinoid receptor agonists and antagonists interact with the dopamine reward pathway and show efficacy in reducing nicotine addiction-like behaviors in preclinical studies. Metformin is currently approved by the Food and Drug Administration for the treatment of diabetes. It activates specific intracellular kinases that may protect against the lower metabolism, higher oxidation, and inflammation that are associated with nicotine withdrawal. Further studies are needed to investigate non-nicotinic targets to improve the treatment of tobacco use disorder. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Lauren C Smith
- Department of Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA; Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Olivier George
- Department of Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA; Department of Psychiatry, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
13
|
Tiwari RK, Sharma V, Pandey RK, Shukla SS. Nicotine Addiction: Neurobiology and Mechanism. J Pharmacopuncture 2020; 23:1-7. [PMID: 32322429 PMCID: PMC7163392 DOI: 10.3831/kpi.2020.23.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/29/2019] [Accepted: 12/16/2019] [Indexed: 01/03/2023] Open
Abstract
Nicotine, primary component of tobaco produces craving and withdrawal effect both in humans and animals. Nicotine shows a close resemblance to other addictive drugs in molecular, neuroanatomical and pharmacological, particularly the drugs which enhances the cognitive functions. Nicotine mainly shows its action through specific nicotinic acetylcholine receptors located in brain. It stimulates presynaptic acetylcholine receptors thereby enhancing Ach release and metabolism. Dopaminergic system is also stimulated by it, thus increasing the concentration of dopamine in nuclear accumbens. This property of nicotine according to various researchers is responsible for reinforcing behavioral change and dependence of nicotine. Various researchers have also depicted that some non dopaminergic systems are also involved for rewarding effect of nicotinic withdrawal. Neurological systems such as GABAergic, serotonergic, noradrenergic, and brain stem cholinergic may also be involved to mediate the actions of nicotine. Further, the neurobiological pathway to nicotine dependence might perhaps be appropriate to the attachment of nicotine to nicotinic acetylcholine receptors, peruse by stimulation of dopaminergic system and activation of general pharmacological changes that might be responsible for nicotine addiction. It is also suggested that MAO A and B both are restrained by nicotine. This enzyme helps in degradation dopamine, which is mainly responsible for nicotinic actions and dependence. Various questions remain uninsurable to nicotine mechanism and require more research. Also, various genetic methods united with modern instrumental analysis might result for more authentic information for nicotine addiction.
Collapse
Affiliation(s)
| | - Vikas Sharma
- Columbia Institute of Pharmacy, Raipur, C.G., India
| | | | | |
Collapse
|
14
|
Wilkerson JL, Deba F, Crowley ML, Hamouda AK, McMahon LR. Advances in the In vitro and In vivo pharmacology of Alpha4beta2 nicotinic receptor positive allosteric modulators. Neuropharmacology 2020; 168:108008. [PMID: 32113032 DOI: 10.1016/j.neuropharm.2020.108008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 11/29/2022]
Abstract
Receptors containing α4 and β2 subunits are a major neuronal nicotinic acetylcholine receptor (nAChR) subtype in the brain. This receptor plays a critical role in nicotine addiction, with potential smoking cessation therapeutics producing modulation of α4β2 nAChR. In addition, compounds that act as agonists at α4β2 nAChR may be useful for the treatment of pathological pain. Further, as the α4β2 nAChR has been implicated in cognition, therapeutics that act as α4β2 nAChR agonists are also being examined as treatments for cognitive disorders and neurological diseases that impact cognitive function, such as Alzheimer's disease and schizophrenia. This review will cover the molecular in vitro evidence that allosteric modulators of the α4β2 neuronal nAChR provide several advantages over traditional α4β2 nAChR orthosteric ligands. Specifically, we explore the concept that nAChR allosteric modulators allow for greater pharmacological selectivity, while minimizing potential deleterious off-target effects. Further, here we discuss the development and preclinical in vivo behavioral assessment of allosteric modulators at the α4β2 neuronal nAChR as therapeutics for smoking cessation, pathological pain, as well as cognitive disorders and neurological diseases that impact cognitive function. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States.
| | - Farah Deba
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Texas at Tyler, Tyler, TX, 75799, United States
| | - Morgan L Crowley
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Ayman K Hamouda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Texas at Tyler, Tyler, TX, 75799, United States.
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States.
| |
Collapse
|
15
|
Activation of GLP-1 receptors attenuates oxycodone taking and seeking without compromising the antinociceptive effects of oxycodone in rats. Neuropsychopharmacology 2020; 45:451-461. [PMID: 31581176 PMCID: PMC6969180 DOI: 10.1038/s41386-019-0531-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022]
Abstract
Despite the effectiveness of current medications to treat opioid use disorder, there is still a high rate of relapse following detoxification. Thus, there is critical need for innovative studies aimed at identifying novel neurobiological mechanisms that could be targeted to treat opioid use disorder. A growing body of preclinical evidence indicates that glucagon-like peptide-1 (GLP-1) receptor agonists reduce drug reinforcement. However, the efficacy of GLP-1 receptor agonists in attenuating opioid-mediated behaviors has not been thoroughly investigated. Using recently established models of opioid-taking and -seeking behaviors, we showed that systemic administration of the GLP-1 receptor agonist exendin-4 reduced oxycodone self-administration and the reinstatement of oxycodone-seeking behavior in rats. We also identified behaviorally selective doses of exendin-4 that reduced opioid-taking and -seeking behaviors and did not produce adverse feeding effects in oxycodone-experienced rats. To identify a central site of action, we showed that systemic exendin-4 penetrated the brain and bound putative GLP-1 receptors on dopamine D1 receptor- and dopamine D2 receptor-expressing medium spiny neurons in the nucleus accumbens shell. Consistent with our systemic studies, infusions of exendin-4 directly into the accumbens shell attenuated oxycodone self-administration and the reinstatement of oxycodone-seeking behavior without affecting ad libitum food intake. Finally, exendin-4 did not alter the analgesic effects of oxycodone, suggesting that activation of GLP-1 receptors attenuated opioid reinforcement without reducing the thermal antinociceptive effects of oxycodone. Taken together, these findings suggest that GLP-1 receptors could serve as potential molecular targets for pharmacotherapies aimed at reducing opioid use disorder.
Collapse
|
16
|
Jackson A, Alkhlaif Y, Papke RL, Brunzell DH, Damaj MI. Impact of modulation of the α7 nicotinic acetylcholine receptor on nicotine reward in the mouse conditioned place preference test. Psychopharmacology (Berl) 2019; 236:3593-3599. [PMID: 31302720 PMCID: PMC6895411 DOI: 10.1007/s00213-019-05331-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/08/2019] [Indexed: 12/26/2022]
Abstract
RATIONALE The α7 nicotinic acetylcholine receptor (nAChR) has been implicated as a target in modulating nicotine reward. However, the effect of pharmacological agents that have been shown to alter the channel properties of the α7 nAChR is not well understood in nicotine reward. OBJECTIVES This study aimed to investigate the impact of α7 nAChR pharmacological modulation on nicotine conditioned place preference (CPP) in mice by using positive allosteric modulators (PAMs) and a silent agonist. METHODS The effect of the orthosteric α7 nAChR full agonist PNU282987 (1.3 and 9 mg/kg, s.c.), Type I α7 PAM NS1738 (1 and 10 mg/kg; i.p.), the Type II α7 PAM PNU120596 (0.3, 1, and 3 mg/kg, i.p.), and the α7 silent agonist NS6740 (1 and 3 mg/kg, i.p) on nicotine CPP was measured in mice. Mice were conditioned with either saline or nicotine (0.5 mg/kg) for 3 days in the CPP paradigm. RESULTS The α7 full orthosteric agonist PNU282987 and the Type II α7 nAChR PAM PNU120596 reduced nicotine CPP, while the silent agonist NS6740 and Type I PAM NS1738 had no effect. The effects of PNU282987 and PNU120596 did not have an effect on morphine CPP. CONCLUSIONS Taken together, our results suggest that modulation of the α7 nAChR can play important roles in nicotine CPP in mice. In addition, the Type II α7 nAChR PAM PNU120596 attenuated nicotine reward suggesting that endogenous acetylcholine/choline tone is sufficient to reduce nicotine CPP. These findings highlight a beneficial effect of using α7 nAChR PAMs in nicotine reward.
Collapse
Affiliation(s)
- Asti Jackson
- Department of Psychiatry, Yale School of Medicine, 34 Park St., New Haven, CT, 06519, USA.
| | - Y. Alkhlaif
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - R. L. Papke
- Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267, Gainesville, FL 32610-0267, USA
| | - D. H. Brunzell
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - M. I. Damaj
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| |
Collapse
|
17
|
Carroll KM, DeVito EE, Yip SW, Nich C, Sofuoglu M. Double‐Blind Placebo‐Controlled Trial of Galantamine for Methadone‐Maintained Individuals With Cocaine Use Disorder: Secondary Analysis of Effects on Illicit Opioid Use. Am J Addict 2019; 28:238-245. [DOI: 10.1111/ajad.12904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/21/2019] [Accepted: 04/21/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
- Kathleen M. Carroll
- Department of Psychiatry, Division on AddictionYale University School of MedicineNew Haven Connecticut
| | - Elise E. DeVito
- Department of Psychiatry, Division on AddictionYale University School of MedicineNew Haven Connecticut
| | - Sarah W. Yip
- Department of Psychiatry, Division on AddictionYale University School of MedicineNew Haven Connecticut
| | - Charla Nich
- Department of Psychiatry, Division on AddictionYale University School of MedicineNew Haven Connecticut
| | - Mehmet Sofuoglu
- Department of Psychiatry, Division on AddictionYale University School of MedicineNew Haven Connecticut
| |
Collapse
|
18
|
Shawky E, El Sohafy SM, de Andrade JP, de Souza Borges W. Profiling of acetylcholinesterase inhibitory alkaloids from some Crinum, Habranthus and Zephyranthes species by GC-MS combined with multivariate analyses and in silico studies. Nat Prod Res 2019; 35:807-814. [PMID: 30990078 DOI: 10.1080/14786419.2019.1598989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Acetylcholinesterase (AChE) inhibitors remain the class of drugs used for the treatment of Alzheimer disease (AD). For the aim of discovering new sources of potent AChE inhibitors, a combined AChE-inhibitory activity together with alkaloid profiles by GC-MS, combined with multivariate statistical analysis for biomarkers determination and in silico studies were attempted. Strategy was applied on leaves, roots and bulbs of six aquatic and terrestrial Amaryllidaceae species. Thirty alkaloids were identified and the AChE inhibitory activities of the extracts were tested by in-vitro Ellman method. Principal bioactive markers were discovered by correlating AChE inhibitory activity with chemical fingerprints via PLS and OPLS modeling which revealed that galanthamine, lycoramine, caranine, tazettine and N-demethylgalanthamine were the most bio-significant markers. Furthermore, the molecular docking was performed to illustrate binding orientations of the top scoring alkaloids in the active site of human acetylcholinesterase. Suggested strategy revealed that, beside galanthamine, caranine, N-demethylgalanthamine, and lycoramine are promising AChE inhibitors.
Collapse
Affiliation(s)
- Eman Shawky
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Samah M El Sohafy
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | | |
Collapse
|
19
|
Moerke MJ, McMahon LR. Nicotine-like discriminative stimulus effects of acetylcholinesterase inhibitors and a muscarinic receptor agonist in Rhesus monkeys. Drug Dev Ind Pharm 2019; 45:861-867. [PMID: 30712397 DOI: 10.1080/03639045.2019.1578787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Acetylcholinesterase (AChE) inhibitors and positive allosteric nicotinic acetylcholine receptor (nAChR) modulators are potential pharmacotherapies for nicotine dependence. Because some smoking cessation aids (e.g. varenicline) appear to work by mimicking the effects of nicotine, we used drug discrimination to examine whether AChE inhibitors and nAChR allosteric modulators mimic the effects of nicotine. Rhesus monkeys discriminated 1.78 mg/kg of nicotine s.c. under an FR5 schedule of stimulus-shock termination. Nicotine and the AChE inhibitors donepezil and galantamine dose-dependently increased responding on the nicotine-appropriate lever with ED50 values of 0.35, 0.22, and 0.77 mg/kg, respectively. Donepezil (0.56 mg/kg) produced nicotine-like effects for at least 6 h, whereas the duration of action of galantamine (1.78 mg/kg) was less than 3 h. The positive allosteric nAChR modulator PNU-120596 (up to 10 mg/kg) and midazolam (up to 1.0 mg/kg) produced no more than 22% nicotine-lever responding. Oxotremorine, a muscarinic acetylcholine receptor agonist that was used to explore the extent to which muscarinic receptor agonism might contribute to the effects of AChE inhibitors, produced 94% nicotine-lever responding (ED50 value 0.013 mg/kg). The muscarinic antagonist atropine significantly antagonized the effects of both oxotremorine and nicotine; however, the dose of atropine antagonizing oxotremorine was smaller than the dose required to antagonize nicotine. Collectively, these results suggest that AChE inhibitors can mimic the effects of nicotine by indirectly stimulating both nicotinic and muscarinic receptors. Inasmuch as some smoking cessation aids work by exerting nicotine-like effects, the current results are consistent with the potential use of AChE inhibitors as novel smoking cessation aids.
Collapse
Affiliation(s)
- Megan J Moerke
- a Department of Pharmacodynamics , College of Pharmacy, University of Florida , Gainesville , FL , USA
| | - Lance R McMahon
- a Department of Pharmacodynamics , College of Pharmacy, University of Florida , Gainesville , FL , USA
| |
Collapse
|
20
|
Schmidt HD, Rupprecht LE, Addy NA. Neurobiological and Neurophysiological Mechanisms Underlying Nicotine Seeking and Smoking Relapse. MOLECULAR NEUROPSYCHIATRY 2019; 4:169-189. [PMID: 30815453 PMCID: PMC6388439 DOI: 10.1159/000494799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022]
Abstract
Tobacco-related morbidity and mortality continue to be a significant public health concern. Unfortunately, current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, a better understanding of the neurobiological and neurophysiological mechanisms that promote smoking relapse is needed to develop novel smoking cessation medications. Here, we review preclinical studies focused on identifying the neurotransmitter and neuromodulator systems that mediate nicotine relapse, often modeled in laboratory animals using the reinstatement paradigm, as well as the plasticity-dependent neurophysiological mechanisms that facilitate nicotine reinstatement. Particular emphasis is placed on how these neuroadaptations relate to smoking relapse in humans. We also highlight a number of important gaps in our understanding of the neural mechanisms underlying nicotine reinstatement and critical future directions, which may lead toward the development of novel, target pharmacotherapies for smoking cessation.
Collapse
Affiliation(s)
- Heath D. Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E. Rupprecht
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
- Interdepartmental Neuroscience Program, Yale Graduate School of Arts and Sciences, New Haven, Connecticut, USA
| |
Collapse
|
21
|
MacLean RR, Waters AJ, Brede E, Sofuoglu M. Effects of galantamine on smoking behavior and cognitive performance in treatment-seeking smokers prior to a quit attempt. Hum Psychopharmacol 2018; 33:e2665. [PMID: 29926988 PMCID: PMC6168949 DOI: 10.1002/hup.2665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/27/2018] [Accepted: 05/18/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Drugs that enhance cholinergic transmission have demonstrated promise treating addictive disorders. Galantamine, an acetylcholinesterase inhibitor, may reduce cigarette smoking in otherwise healthy treatment-seeking smokers. METHODS The current study is a double-blind, placebo-controlled, study that randomized daily smokers (n = 60) to receive one of two doses of galantamine extended release (8 or 16 mg/day), or a placebo treatment. Participants completed a smoking choice task as well as study measures and cognitive tasks in the laboratory and daily life using ecological momentary assessment. Analysis focused on smoking behavior and satisfaction, cognitive performance, and decision to smoke prior to a quit attempt. RESULTS Linear mixed models demonstrated that, compared with placebo, both doses of galantamine reduced smoking in a laboratory choice task (p = 0.006) and decreased urine cotinine levels, but not self-reported cigarettes, during the pre-quit period (p = 0.007). Treatment had minimal effect on smoking satisfaction or cognitive performance. CONCLUSIONS The results suggest that galantamine reduces nicotine intake but it is unlikely that galantamine improves cognitive performance in otherwise healthy, treatment-seeking smokers. Larger randomized clinical trials can determine if galantamine adjunctive to addiction treatment can improve smoking treatment outcomes.
Collapse
Affiliation(s)
- Robert Ross MacLean
- VA Connecticut Healthcare System, West Haven, Connecticut,Yale University School of Medicine, New Haven, Connecticut
| | - Andrew J. Waters
- Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Emily Brede
- National Institute of Health, Bethesda, Maryland
| | - Mehmet Sofuoglu
- VA Connecticut Healthcare System, West Haven, Connecticut,Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
22
|
Perkins KA, Roy Chengappa KN, Karelitz JL, Boldry MC, Michael V, Herb T, Gannon J, Brar J, Ford L, Rassnick S, Brunzell DH. Initial Cross-Over Test of A Positive Allosteric Modulator of Alpha-7 Nicotinic Receptors to Aid Cessation in Smokers With Or Without Schizophrenia. Neuropsychopharmacology 2018; 43:1334-1342. [PMID: 29185480 PMCID: PMC5916362 DOI: 10.1038/npp.2017.292] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/20/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022]
Abstract
Preclinical research shows that compounds acting at α7 nicotinic receptors (nAChRs) can reduce nicotine self-administration, suggesting that a positive allosteric modulator (PAM) of α7 receptors, JNJ-39393406, may aid smoking cessation. Moreover, individuals with schizophrenia, who have very high rates of smoking, have reduced expression of α7 nAChRs and may particularly benefit from this compound. In two parallel studies using a within-subject cross-over design, 36 healthy smokers (Study 1) and 62 smokers with schizophrenia (Study 2), both groups high in quit interest, attempted to initiate quitting temporarily during each of two 3-week phases. Treatments were the α7 nicotinic receptor PAM JNJ-39393406 (100 mg b.i.d.) or placebo (double-blind, counter-balanced). In each phase, all smoked ad lib with no drug on week 1 or during dose run-up on week 2, and then tried to quit every day during week 3. Abstinence (confirmed by CO <5 p.p.m.) and smoking reduction (CO <8), as well as cigarettes/day (in Study 1), were assessed daily (Monday-Friday) each quit week and compared between conditions. Secondary outcomes included abstinence symptoms (withdrawal and craving) and cognitive test responding (N-back; continuous performance task). In both studies, compared with placebo, active JNJ-39393406 did not increase the number of abstinent days nor reduce total smoking exposure. We also found no significant improvements in craving, withdrawal, or cognitive function. With this dose and study duration, our findings do not support further testing of this α7 nAChR PAM compound for possible efficacy in smoking cessation, in smokers with or without schizophrenia.
Collapse
Affiliation(s)
- Kenneth A Perkins
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA,Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, 3811 O’Hara Street, Pittsburgh, PA 15213, USA, Tel: +1 412 246 5395, Fax: +1 412 246 5390, E-mail:
| | - K N Roy Chengappa
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Joshua L Karelitz
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Margaret C Boldry
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Valerie Michael
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Taylor Herb
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Jessica Gannon
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Jaspreet Brar
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Lisa Ford
- Janssen Research and Development, Janssen Pharmaceuticals, Titusville NJ, USA
| | - Stefanie Rassnick
- Janssen Research and Development, Janssen Pharmaceuticals, Titusville NJ, USA
| | - Darlene H Brunzell
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond VA, USA
| |
Collapse
|
23
|
Altaf AA, Hamayun M, Lal B, Tahir MN, Holder AA, Badshah A, Crans DC. Ferrocene-based anilides: synthesis, structural characterization and inhibition of butyrylcholinesterase. Dalton Trans 2018; 47:11769-11781. [DOI: 10.1039/c8dt01726c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Twenty-three compounds in two series of ferrocene-based anilides, with the general formula C5H5-Fe-C5H4-C6H4-NH-CO-C6H4-R (where R = H, F, Cl, CH3 and OCH3), have been synthesized and found to inhibit butyrylcholinesterase.
Collapse
Affiliation(s)
- Ataf Ali Altaf
- Department of Chemistry
- University of Gujrat
- Gujrat 50700
- Pakistan
| | | | - Bhajan Lal
- Department of Chemistry
- Shah Abdul Latif University
- Khairpur
- Pakistan
| | | | - Alvin A. Holder
- Department of Chemistry and Biochemistry
- Old Dominion University
- Norfolk
- USA
| | - Amin Badshah
- Department of Chemistry
- Quaid-i-Azam University
- Islamabad-45320
- Pakistan
| | - Debbie C. Crans
- Department of Chemistry
- Colorado State University
- Fort Collins
- USA
| |
Collapse
|
24
|
Moro F, Orrù A, Marzo CM, Di Clemente A, Cervo L. mGluR2/3 mediates short-term control of nicotine-seeking by acute systemic N-acetylcysteine. Addict Biol 2018; 23:28-40. [PMID: 27558879 DOI: 10.1111/adb.12443] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/24/2016] [Accepted: 08/01/2016] [Indexed: 12/26/2022]
Abstract
Chronic self-administration of nicotine induces maladaptive changes in the cortico-accumbal glutamate (Glu) network. Consequently, re-exposure to nicotine-associated cues raises extracellular Glu in the nucleus accumbens reinstating drug-seeking. Restoring basal concentrations of extracellular Glu, thereby increasing tonic activation of the presynaptic group II metabotropic Glu receptors (mGluR2/3) with N-acetylcysteine (N-AC), might offer a valid therapeutic approach for maintaining smoking abstinence. Although N-AC modulates nicotine-seeking behavior by drug-associated stimuli in abstinent rats, it is still unclear whether it occurs through activation of mGluR2/3. Male Wistar rats were trained to associate discriminative stimuli (SD s) with the availability of intravenous nicotine (0.03 mg/kg/65 µl/2-second/infusion) or oral saccharin (100 µl of 50 mg/l) self-administration versus non-reward. Reinforced response was followed by a cue signaling 20-second time-out (CSs). Once the training criterion was met, rats underwent lever press extinction, without reinforcers, SD s and CSs. Re-exposure to nicotine or saccharin SD+ /CS+ , but not non-reward SD- /CS- , revived responding on the previously reinforced lever. Acute N-AC, 100 but not 60 or 30 mg/kg i.p., reduced cue-induced nicotine-seeking. N-AC 100 mg/kg did not modify cue-induced saccharin-seeking behavior or influenced locomotor activity. Blocking mGluR2/3 with the selective antagonist LY341495, 1 mg/kg i.p., completely prevented the antirelapse activity of N-AC. The finding that N-AC prevents cue-induced nicotine-seeking by stimulating mGluR2/3 might indicate a therapeutic opportunity for acute cue-controlled nicotine-seeking. Future studies could evaluate the persistent effects of chronic N-AC in promoting enduring suppression of nicotine-cue conditioned responding.
Collapse
Affiliation(s)
- Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Alessandro Orrù
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Claudio Marcello Marzo
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| |
Collapse
|
25
|
Wu J, Cippitelli A, Zhang Y, Debevec G, Schoch J, Ozawa A, Yu Y, Liu H, Chen W, Houghten RA, Welmaker GS, Giulianotti MA, Toll L. Highly Selective and Potent α4β2 nAChR Antagonist Inhibits Nicotine Self-Administration and Reinstatement in Rats. J Med Chem 2017; 60:10092-10104. [PMID: 29178785 DOI: 10.1021/acs.jmedchem.7b01250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The α4β2 nAChR is the most predominant subtype in the brain and is a well-known culprit for nicotine addiction. Previously we presented a series of α4β2 nAChR selective compounds that were discovered from a mixture-based positional-scanning combinatorial library. Here we report further optimization identified highly potent and selective α4β2 nAChR antagonists 5 (AP-202) and 13 (AP-211). Both compounds are devoid of in vitro agonist activity and are potent inhibitors of epibatidine-induced changes in membrane potential in cells containing α4β2 nAChR, with IC50 values of approximately 10 nM, but are weak agonists in cells containing α3β4 nAChR. In vivo studies show that 5 can significantly reduce operant nicotine self-administration and nicotine relapse-like behavior in rats at doses of 0.3 and 1 mg/kg. The pharmacokinetic data also indicate that 5, via sc administration, is rapidly absorbed into the blood, reaching maximal concentration within 10 min with a half-life of less than 1 h.
Collapse
Affiliation(s)
- Jinhua Wu
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Assuage Pharmaceuticals, Inc , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Andrea Cippitelli
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Yaohong Zhang
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Institute of Materia Medica, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, P. R. China.,School of Chemistry and Chemical Engineering, Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University , Shaoxing 312000, Zhejiang, P. R. China
| | - Ginamarie Debevec
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Jennifer Schoch
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Akihiko Ozawa
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Yongping Yu
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Institute of Materia Medica, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, P. R. China
| | - Huan Liu
- Institute of Materia Medica, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, P. R. China
| | - Wenteng Chen
- Institute of Materia Medica, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, P. R. China
| | - Richard A Houghten
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Assuage Pharmaceuticals, Inc , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Gregory S Welmaker
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Assuage Pharmaceuticals, Inc , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Marc A Giulianotti
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Assuage Pharmaceuticals, Inc , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Lawrence Toll
- Torrey Pines Institute for Molecular Studies , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States.,Assuage Pharmaceuticals, Inc , 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| |
Collapse
|
26
|
Reiner DJ, Mietlicki-Baase EG, Olivos DR, McGrath LE, Zimmer DJ, Koch-Laskowski K, Krawczyk J, Turner CA, Noble EE, Hahn JD, Schmidt HD, Kanoski SE, Hayes MR. Amylin Acts in the Lateral Dorsal Tegmental Nucleus to Regulate Energy Balance Through Gamma-Aminobutyric Acid Signaling. Biol Psychiatry 2017; 82:828-838. [PMID: 28237459 PMCID: PMC5503810 DOI: 10.1016/j.biopsych.2016.12.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/13/2016] [Accepted: 12/28/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pancreatic- and brain-derived hormone amylin promotes negative energy balance and is receiving increasing attention as a promising obesity therapeutic. However, the neurobiological substrates mediating amylin's effects are not fully characterized. We postulated that amylin acts in the lateral dorsal tegmental nucleus (LDTg), an understudied neural processing hub for reward and homeostatic feeding signals. METHODS We used immunohistochemical and quantitative polymerase chain reaction analyses to examine expression of the amylin receptor complex in rat LDTg tissue. Behavioral experiments were performed to examine the mechanisms underlying the hypophagic effects of amylin receptor activation in the LDTg. RESULTS Immunohistochemical and quantitative polymerase chain reaction analyses show expression of the amylin receptor complex in the LDTg. Activation of LDTg amylin receptors by the agonist salmon calcitonin dose-dependently reduces body weight, food intake, and motivated feeding behaviors. Acute pharmacological studies and longer-term adeno-associated viral knockdown experiments indicate that LDTg amylin receptor signaling is physiologically and potentially preclinically relevant for energy balance control. Finally, immunohistochemical data indicate that LDTg amylin receptors are expressed on gamma-aminobutyric acidergic neurons, and behavioral results suggest that local gamma-aminobutyric acid receptor signaling mediates the hypophagia after LDTg amylin receptor activation. CONCLUSIONS These findings identify the LDTg as a novel nucleus with therapeutic potential in mediating amylin's effects on energy balance through gamma-aminobutyric acid receptor signaling.
Collapse
Affiliation(s)
- David J Reiner
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth G Mietlicki-Baase
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Diana R Olivos
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Lauren E McGrath
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Derek J Zimmer
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kieran Koch-Laskowski
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joanna Krawczyk
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christopher A Turner
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily E Noble
- Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, California
| | - Joel D Hahn
- Neurobiology Section, University of Southern California, Los Angeles, California
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, California
| | - Matthew R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
27
|
Altaf AA, Kausar S, Hamayun M, Lal B, Tahir MN, Badshah A. Ferrocenylaniline based amide analogs of methoxybenzoic acids: Synthesis, structural characterization and butyrylcholinesterase (BChE) inhibition studies. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.05.132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
28
|
Bruijnzeel AW. Neuropeptide systems and new treatments for nicotine addiction. Psychopharmacology (Berl) 2017; 234:1419-1437. [PMID: 28028605 PMCID: PMC5420481 DOI: 10.1007/s00213-016-4513-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE The mildly euphoric and cognitive enhancing effects of nicotine play a role in the initiation of smoking, while dysphoria and anxiety associated with smoking cessation contribute to relapse. After the acute withdrawal phase, smoking cues, a few cigarettes (i.e., lapse), and stressors can cause relapse. Human and animal studies have shown that neuropeptides play a critical role in nicotine addiction. OBJECTIVES The goal of this paper is to describe the role of neuropeptide systems in the initiation of nicotine intake, nicotine withdrawal, and the reinstatement of extinguished nicotine seeking. RESULTS The reviewed studies indicate that several drugs that target neuropeptide systems diminish the rewarding effects of nicotine by preventing the activation of dopaminergic systems. Other peptide-based drugs diminish the hyperactivity of brain stress systems and diminish withdrawal-associated symptom severity. Blockade of hypocretin-1 and nociceptin receptors and stimulation of galanin and neurotensin receptors diminishes the rewarding effects of nicotine. Both corticotropin-releasing factor type 1 and kappa-opioid receptor antagonists diminish dysphoria and anxiety-like behavior associated with nicotine withdrawal and inhibit stress-induced reinstatement of nicotine seeking. Furthermore, blockade of vasopressin 1b receptors diminishes dysphoria during nicotine withdrawal, and melanocortin 4 receptor blockade prevents stress-induced reinstatement of nicotine seeking. The role of neuropeptide systems in nicotine-primed and cue-induced reinstatement is largely unexplored, but there is evidence for a role of hypocretin-1 receptors in cue-induced reinstatement of nicotine seeking. CONCLUSION Drugs that target neuropeptide systems might decrease the euphoric effects of smoking and improve relapse rates by diminishing withdrawal symptoms and improving stress resilience.
Collapse
Affiliation(s)
- Adriaan W. Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, Florida, USA,Department of Neuroscience, University of Florida, Gainesville, Florida, USA,Center for Addiction Research and Education, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
29
|
Attenuation of nicotine taking and seeking in rats by the stoichiometry-selective alpha4beta2 nicotinic acetylcholine receptor positive allosteric modulator NS9283. Psychopharmacology (Berl) 2017; 234:475-484. [PMID: 27844094 DOI: 10.1007/s00213-016-4475-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/02/2016] [Indexed: 12/16/2022]
Abstract
RATIONALE The rewarding and reinforcing effects of nicotine are produced, in large part, by activation of neuronal α4β2* nicotinic acetylcholine receptors (nAChRs), pentameric protein complexes comprised of different stoichiometries of α4 and β2 subunits. However, little is known about the functional role of distinct subtypes of α4β2* nAChRs in nicotine addiction. OBJECTIVES NS9283 represents a new class of stoichiometry-selective positive allosteric modulators (PAMs) that selectively bind to α4β2 nAChRs containing three α4 and two β2 subunits (3(α4)2(β2) nAChRs). The present experiments were designed to determine the effects of NS9283 on nicotine self-administration and the reinstatement of nicotine-seeking behavior, an animal model of smoking relapse. Parallel studies of sucrose self-administration and reinstatement were conducted in separate cohorts of rats to determine if the effects of NS9283 generalized to other reinforced behaviors. RESULTS Acute and repeated administration of NS9283 dose-dependently reduced nicotine self-administration and reinstatement in male Sprague Dawley rats. These effects were reinforcer specific as no effects of NS9283 on sucrose self-administration and reinstatement were noted. NS9283 also failed to substitute for nicotine in supporting self-administration behavior suggesting that, at the doses tested, NS9283 alone is not reinforcing. CONCLUSION Taken together, these results provide compelling evidence that stoichiometry-selective PAMs of 3(α4)2(β2) nAChRs attenuate nicotine taking and seeking in rats and suggest that targeting 3(α4)2(β2) nAChRs may represent a promising therapeutic strategy for preventing smoking relapse.
Collapse
|
30
|
A novel one-step electrochemical preparation of silver nanoparticles/poly(3-methylthiophene) nanocomposite for detection of galantamine in human cerebrospinal fluid and narcissus. J Electroanal Chem (Lausanne) 2017. [DOI: 10.1016/j.jelechem.2016.12.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
31
|
Biswas L, Harrison E, Gong Y, Avusula R, Lee J, Zhang M, Rousselle T, Lage J, Liu X. Enhancing effect of menthol on nicotine self-administration in rats. Psychopharmacology (Berl) 2016; 233:3417-27. [PMID: 27473365 PMCID: PMC4990499 DOI: 10.1007/s00213-016-4391-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/08/2016] [Indexed: 01/19/2023]
Abstract
RATIONALE Tobacco smoking is a leading preventable cause of premature death in the USA. Menthol is a significant flavoring additive in tobacco products. Clinical evidence suggests that menthol may promote tobacco smoking and nicotine dependence. However, it is unclear whether menthol enhances the reinforcing actions of nicotine and thus facilitates nicotine consumption. This study employed a rat model of nicotine self-administration to examine the effects of menthol on nicotine-taking behavior. METHODS Male Sprague-Dawley rats were trained in daily 1-h sessions to press a lever for intravenous nicotine self-administration under a fixed-ratio 5 schedule of reinforcement. In separate groups, rats self-administered nicotine at four different doses (0.0075, 0.015, 0.03, and 0.06 mg/kg/infusion). Five minutes prior to the two test sessions, menthol (5 mg/kg) or its vehicle was administered intraperitoneally in all rats in a counterbalanced design within each group. In separate rats that self-administered 0.015 mg/kg/infusion nicotine, menthol dose-response function was determined. Menthol was also tested on food self-administration. RESULTS An inverted U-shaped nicotine dose-response curve was observed. Menthol pretreatment shifted the nicotine dose-response curve to the left. The facilitating effect of menthol on the self-administration of 0.015 mg/kg/infusion nicotine was dose-dependent, whereas it produced similar effects at doses above the threshold of 2.5 mg/kg. Menthol tended to suppress the self-administration of food pellets. CONCLUSIONS These data demonstrate that menthol enhances the reinforcing effects of nicotine, and the effect of menthol was specific to nicotine. The findings suggest that menthol directly facilitates nicotine consumption, thereby contributing to tobacco smoking.
Collapse
Affiliation(s)
- Lisa Biswas
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Erin Harrison
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Yongzhen Gong
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Ramachandram Avusula
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Jonathan Lee
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Meiyu Zhang
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Thomas Rousselle
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Janice Lage
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Xiu Liu
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
32
|
Abstract
Natural rewards, including food, water, sleep and social interactions, are required to sustain life. The neural substrates that regulate the reinforcing effects of these behaviors are also the same neurobiological mechanisms mediating mood, motivation and the rewarding effects of pharmacological stimuli. That the neuropeptide glucagon-like peptide-1 (GLP-1) is under investigation for both the homeostatic and hedonic controls of feeding is not surprising or novel. However, if the neural substrates that underline food reward are shared with other reward-related behaviors generally, then future research should investigate and embrace the likelihood that endogenous and exogenous GLP-1 receptor activation may influence multiple reward-related behaviors. Indeed, studies of the neurobiological mechanisms underlying motivated feeding behavior have informed much of the basic research investigating neural substrates of drug addiction. An emerging literature demonstrates a role for the GLP-1 system in modulating maladaptive reward behaviors, including drug and alcohol consumption. Thus, if GLP-1-based pharmacotherapies are to be used to treat drug addiction and other diseases associated with maladaptive reward behaviors (e.g. obesity and eating disorders), the neuroscience field must conduct systematic, mechanistic neuropharmacological and behavioral studies of each GLP-1 receptor-expressing nucleus within the brain. It is possible that behavioral selectivity may result from these studies, which could inform future approaches to targeting GLP-1R signaling in discrete brain nuclei to treat motivated behaviors. Equally as likely, non-selective effects on natural reward and maladaptive reward behaviors may be observed for GLP-1-based pharmacotherapies. In this case, a better understanding of the effects of increased central GLP-1R activation on motivated behaviors will aid in clinical approaches toward treating aberrant feeding behaviors and/or drug dependence.
Collapse
Affiliation(s)
- Matthew R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania
| |
Collapse
|
33
|
Zhou X, Li Y, Shi X, Ma C. An overview on therapeutics attenuating amyloid β level in Alzheimer's disease: targeting neurotransmission, inflammation, oxidative stress and enhanced cholesterol levels. Am J Transl Res 2016; 8:246-69. [PMID: 27158324 PMCID: PMC4846881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 06/05/2023]
Abstract
Alzheimer's disease (AD) is the most common underlying cause of dementia, and novel drugs for its treatment are needed. Of the different theories explaining the development and progression of AD, "amyloid hypothesis" is the most supported by experimental data. This hypothesis states that the cleavage of amyloid precursor protein (APP) leads to the formation of amyloid beta (Aβ) peptides that congregate with formation and deposition of Aβ plaques in the frontal cortex and hippocampus. Risk factors including neurotransmitter modulation, chronic inflammation, metal-induced oxidative stress and elevated cholesterol levels are key contributors to the disease progress. Current therapeutic strategies abating AD progression are primarily based on anti-acetylcholinesterase (AChE) inhibitors as cognitive enhancers. The AChE inhibitor, donepezil, is proven to strengthen cognitive functions and appears effective in treating moderate to severe AD patients. N-Methyl-D-aspartate receptor antagonist, memantine, is also useful, and its combination with donepezil demonstrated a strong stabilizing effect in clinical studies on AD. Nonsteroidal anti-inflammatory drugs delayed the onset and progression of AD and attenuated cognitive dysfunction. Based upon epidemiological evidence and animal studies, antioxidants emerged as potential AD preventive agents; however, clinical trials revealed inconsistencies. Pharmacokinetic and pharmacodynamic profiling demonstrated pleiotropic functions of the hypolipidemic class of drugs, statins, potentially contributing towards the prevention of AD. In addition, targeting the APP processing pathways, stimulating neuroprotective signaling mechanisms, using the amyloid anti-aggregants and Aβ immunotherapy surfaced as well-tested strategies in reducing the AD-like pathology. Overall, this review covers mechanism of inducing the Aβ formation, key risk factors and major therapeutics prevalent in the AD treatment nowadays. It also delineates the need for novel screening approaches towards identifying drugs that may prevent or at least limit the progression of this devastating disease.
Collapse
Affiliation(s)
- Xiaoling Zhou
- The Affiliated Hospital to Changchun University of Chinese Medicine Changchun, China
| | - Yifei Li
- The Affiliated Hospital to Changchun University of Chinese Medicine Changchun, China
| | - Xiaozhe Shi
- The Affiliated Hospital to Changchun University of Chinese Medicine Changchun, China
| | - Chun Ma
- The Affiliated Hospital to Changchun University of Chinese Medicine Changchun, China
| |
Collapse
|
34
|
Ashare RL, Kimmey BA, Rupprecht LE, Bowers ME, Hayes MR, Schmidt HD. Repeated administration of an acetylcholinesterase inhibitor attenuates nicotine taking in rats and smoking behavior in human smokers. Transl Psychiatry 2016; 6:e713. [PMID: 26784967 PMCID: PMC5068882 DOI: 10.1038/tp.2015.209] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/08/2015] [Accepted: 10/30/2015] [Indexed: 01/14/2023] Open
Abstract
Tobacco smoking remains the leading cause of preventable death worldwide and current smoking cessation medications have limited efficacy. Thus, there is a clear need for translational research focused on identifying novel pharmacotherapies for nicotine addiction. Our previous studies demonstrated that acute administration of an acetylcholinesterase inhibitor (AChEI) attenuates nicotine taking and seeking in rats and suggest that AChEIs could be repurposed for smoking cessation. Here, we expand upon these findings with experiments designed to determine the effects of repeated AChEI administration on voluntary nicotine taking in rats as well as smoking behavior in human smokers. Rats were trained to self-administer intravenous infusions of nicotine (0.03 mg kg(-1) per 0.59 ml) on a fixed-ratio-5 schedule of reinforcement. Once rats maintained stable nicotine taking, galantamine or donepezil was administered before 10 consecutive daily nicotine self-administration sessions. Repeated administration of 5.0 mg kg(-1) galantamine and 3.0 mg kg(-1) donepezil attenuated nicotine self-administration in rats. These effects were reinforcer-specific and not due to adverse malaise-like effects of drug treatment as repeated galantamine and donepezil administration had no effects on sucrose self-administration, ad libitum food intake and pica. The effects of repeated galantamine (versus placebo) on cigarette smoking were also tested in human treatment-seeking smokers. Two weeks of daily galantamine treatment (8.0 mg (week 1) and 16.0 mg (week 2)) significantly reduced smoking rate as well as smoking satisfaction and reward compared with placebo. This translational study indicates that repeated AChEI administration reduces nicotine reinforcement in rats and smoking behavior in humans at doses not associated with tolerance and/or adverse effects.
Collapse
Affiliation(s)
- R L Ashare
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B A Kimmey
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L E Rupprecht
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M E Bowers
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - H D Schmidt
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
35
|
Neuroscience of nicotine for addiction medicine: novel targets for smoking cessation medications. PROGRESS IN BRAIN RESEARCH 2015; 223:191-214. [PMID: 26806777 DOI: 10.1016/bs.pbr.2015.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Morbidity and mortality associated with tobacco smoking constitutes a significant burden on healthcare budgets all over the world. Therefore, promoting smoking cessation is an important goal of health professionals and policy makers throughout the world. Nicotine is a major psychoactive component in tobacco that is largely responsible for the widespread addiction to tobacco. A majority of the currently available FDA-approved smoking cessation medications act via neuronal nicotinic receptors. These medications are effective in approximately half of all the smokers, who want to quit and relapse among abstinent smokers continues to be high. In addition to relapse among abstinent smokers, unpleasant effects associated with nicotine withdrawal are a major motivational factor in continued tobacco smoking. Over the last two decades, animal studies have helped in identifying several neural substrates that are involved in nicotine-dependent behaviors including those associated with nicotine withdrawal and relapse to tobacco smoking. In this review, first the role of specific brain regions/circuits that are involved in nicotine dependence will be discussed. Next, the review will describe the role of specific nicotinic receptor subunits in nicotine dependence. Finally, the review will discuss the role of classical neurotransmitters (dopamine, serotonin, noradrenaline, glutamate, and γ-aminobutyric acid) as well as endogenous opioid and endocannabinoid signaling in nicotine dependence. The nicotinic and nonnicotinic neural substrates involved in nicotine-dependent behaviors can serve as possible targets for future smoking cessation medications.
Collapse
|
36
|
Muscarinic and nicotinic cholinergic receptor antagonists differentially mediate acquisition of fructose-conditioned flavor preference and quinine-conditioned flavor avoidance in rats. Neurobiol Learn Mem 2015; 123:239-49. [DOI: 10.1016/j.nlm.2015.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/16/2015] [Accepted: 07/02/2015] [Indexed: 01/06/2023]
|
37
|
Abstract
The aim of Addiction Biology is to advance our understanding of the action of drugs of abuse and addictive processes via the publication of high-impact clinical and pre-clinical findings resulting from behavioral, molecular, genetic, biochemical, neurobiological and pharmacological research. As of 2013, Addiction Biology is ranked number 1 in the category of Substance Abuse journals (SCI). Occasionally, Addiction Biology likes to highlight via review important findings focused on a particular topic and recently published in the journal. The current review summarizes a number of key publications from Addiction Biology that have contributed to the current knowledge of nicotine research, comprising a wide spectrum of approaches, both clinical and pre-clinical, at the cellular, molecular, systems and behavioral levels. A number of findings from human studies have identified, using imaging techniques, alterations in common brain circuits, as well as morphological and network activity changes, associated with tobacco use. Furthermore, both clinical and pre-clinical studies have characterized a number of mechanistic targets critical to understanding the effects of nicotine and tobacco addiction. Together, these findings will undoubtedly drive future studies examining the dramatic impact of tobacco use and the development of treatments to counter nicotine dependence.
Collapse
Affiliation(s)
- Rick E. Bernardi
- Institute of Psychopharmacology; Central Institute of Mental Health; Medical Faculty Mannheim/Heidelberg University; Germany
| |
Collapse
|
38
|
Rahman S, Engleman EA, Bell RL. Nicotinic receptor modulation to treat alcohol and drug dependence. Front Neurosci 2015; 8:426. [PMID: 25642160 PMCID: PMC4295535 DOI: 10.3389/fnins.2014.00426] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/04/2014] [Indexed: 01/10/2023] Open
Abstract
Alcohol and drug dependence are serious public health problems worldwide. The prevalence of alcohol and drug dependence in the United States and other parts of the world is significant. Given the limitations in the efficacy of current pharmacotherapies to treat these disorders, research in developing alternative pharmacotherapies continues. Preclinical and clinical evidence thus far has indicated that brain nicotinic acetylcholine receptors (nAChRs) are important pharmacological targets for the development of medications to treat alcohol and drug dependence. The nAChRs are a super family of ligand gated ion channels, and are expressed throughout the brain with twelve neuronal nAChR subunits (α2–α10 and β2–β4) identified. Here, we review preclinical and clinical evidence involving a number of nAChR ligands that target different nAChR subtypes in alcohol and nicotine addiction. The important ligands include cytisine, lobeline, mecamylamine, varenicline, sazetidine A and others that target α4β2* nAChR subtypes as small molecule modulators of the brain nicotinic cholinergic system are also discussed. Taken together, both preclinical and clinical data exist that support nAChR–based ligands as promising therapeutic agents for the treatment of alcohol and drug dependence.
Collapse
Affiliation(s)
- Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University Brookings, SD, USA
| | - Eric A Engleman
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine Indianapolis, IN, USA
| | - Richard L Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine Indianapolis, IN, USA
| |
Collapse
|
39
|
Rahman S, Engleman EA, Bell RL. Nicotinic receptor modulation to treat alcohol and drug dependence. Front Neurosci 2015. [DOI: https://doi.org/10.3389/fnins.2014.00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
40
|
Abstract
Nicotine dependence is a chronic, relapsing disorder with complex biological mechanisms underlying the motivational basis for this behavior. Although more than 70 % of current smokers express a desire to quit, most relapse within one year, underscoring the need for novel treatments. A key focus of translational research models addressing nicotine dependence has been on cross-validation of human and animal models in order to improve the predictive value of medication screening paradigms. In this chapter, we review several lines of research highlighting the utility of cross-validation models in elucidating the biological underpinnings of nicotine reward and reinforcement, identifying factors which may influence individual response to treatment, and facilitating rapid translation of findings to practice.
Collapse
|
41
|
Neurobiological Bases of Cue- and Nicotine-induced Reinstatement of Nicotine Seeking: Implications for the Development of Smoking Cessation Medications. Curr Top Behav Neurosci 2015; 24:125-54. [PMID: 25638336 DOI: 10.1007/978-3-319-13482-6_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A better understanding of the neurobiological factors that contribute to relapse to smoking is needed for the development of efficacious smoking cessation medications. Reinstatement procedures allow the preclinical assessment of several factors that contribute to relapse in humans, including re-exposure to nicotine via tobacco smoking and the presentation of stimuli that were previously associated with nicotine administration (i.e., conditioned stimuli). This review provides an integrated discussion of the results of animal studies that used reinstatement procedures to assess the efficacy of pharmacologically targeting various neurotransmitter systems in attenuating the cue- and nicotine-induced reinstatement of nicotine seeking. The results of these animal studies have increased our understanding of the neurobiological processes that mediate the conditioned effects of stimuli that trigger reinstatement to nicotine seeking. Thus, these findings provide important insights into the neurobiological substrates that modulate relapse to tobacco smoking in humans and the ongoing search for novel efficacious smoking cessation medications.
Collapse
|
42
|
Perry CJ, Zbukvic I, Kim JH, Lawrence AJ. Role of cues and contexts on drug-seeking behaviour. Br J Pharmacol 2014; 171:4636-72. [PMID: 24749941 PMCID: PMC4209936 DOI: 10.1111/bph.12735] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/04/2014] [Accepted: 04/10/2014] [Indexed: 01/15/2023] Open
Abstract
Environmental stimuli are powerful mediators of craving and relapse in substance-abuse disorders. This review examined how animal models have been used to investigate the cognitive mechanisms through which cues are able to affect drug-seeking behaviour. We address how animal models can describe the way drug-associated cues come to facilitate the development and persistence of drug taking, as well as how these cues are critical to the tendency to relapse that characterizes substance-abuse disorders. Drug-associated cues acquire properties of conditioned reinforcement, incentive motivation and discriminative control, which allow them to influence drug-seeking behaviour. Using these models, researchers have been able to investigate the pharmacology subserving the behavioural impact of environmental stimuli, some of which we highlight. Subsequently, we examine whether the impact of drug-associated stimuli can be attenuated via a process of extinction, and how this question is addressed in the laboratory. We discuss how preclinical research has been translated into behavioural therapies targeting substance abuse, as well as highlight potential developments to therapies that might produce more enduring changes in behaviour.
Collapse
Affiliation(s)
- Christina J Perry
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, Vic., Australia
- Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, Vic., Australia
| | - Isabel Zbukvic
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, Vic., Australia
- Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, Vic., Australia
| | - Jee Hyun Kim
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, Vic., Australia
- Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, Vic., Australia
| | - Andrew J Lawrence
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, Vic., Australia
- Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, Vic., Australia
| |
Collapse
|
43
|
Alger BE, Nagode DA, Tang AH. Muscarinic cholinergic receptors modulate inhibitory synaptic rhythms in hippocampus and neocortex. Front Synaptic Neurosci 2014; 6:18. [PMID: 25249974 PMCID: PMC4155787 DOI: 10.3389/fnsyn.2014.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/29/2014] [Indexed: 01/08/2023] Open
Abstract
Activation of muscarinic acetylcholine (ACh) receptors (mAChRs) powerfully affects many neuronal properties as well as numerous cognitive behaviors. Small neuronal circuits constitute an intermediate level of organization between neurons and behaviors, and mAChRs affect interactions among cells that compose these circuits. Circuit activity is often assessed by extracellular recordings of the local field potentials (LFPs), which are analogous to in vivo EEGs, generated by coordinated neuronal interactions. Coherent forms of physiologically relevant circuit activity manifest themselves as rhythmic oscillations in the LFPs. Frequencies of rhythmic oscillations that are most closely associated with animal behavior are in the range of 4–80 Hz, which is subdivided into theta (4–14 Hz), beta (15–29 Hz) and gamma (30–80 Hz) bands. Activation of mAChRs triggers rhythmic oscillations in these bands in the hippocampus and neocortex. Inhibitory responses mediated by GABAergic interneurons constitute a prominent feature of these oscillations, and indeed, appear to be their major underlying factor in many cases. An important issue is which interneurons are involved in rhythm generation. Besides affecting cellular and network properties directly, mAChRs can cause the mobilization of endogenous cannabinoids (endocannabinoids, eCBs) that, by acting on the principal cannabinoid receptor of the brain, CB1R, regulate the release of certain neurotransmitters, including GABA. CB1Rs are heavily expressed on only a subset of interneurons and, at lower density, on glutamatergic neurons. Exogenous cannabinoids typically disrupt oscillations in the theta (θ) and gamma (γ) ranges, which probably contributes to the behavioral effects of these drugs. It is important to understand how neuronal circuit activity is affected by mAChR-driven eCBs, as this information will provide deeper insight into the actions of ACh itself, as well as into the effects of eCBs and exogenous cannabinoids in animal behavior. After covering some basic aspects of the mAChR system, this review will focus on recent findings concerning the mechanisms and circuitry that generate θ and γ rhythms in hippocampus and neocortex. The ability of optogenetic methods to probe the many roles of ACh in rhythm generation is highlighted.
Collapse
Affiliation(s)
- Bradley E Alger
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA ; Department of Psychiatry, University of Maryland School of Medicine Baltimore, MD, USA ; Program in Neuroscience, Graduate School, University of Maryland Baltimore Baltimore, MD, USA
| | - Daniel A Nagode
- Department of Biology, University of Maryland College Park College Park, MD, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
44
|
Lee AM, Arreola AC, Kimmey BA, Schmidt HD. Administration of the nicotinic acetylcholine receptor agonists ABT-089 and ABT-107 attenuates the reinstatement of nicotine-seeking behavior in rats. Behav Brain Res 2014; 274:168-75. [PMID: 25128791 DOI: 10.1016/j.bbr.2014.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/04/2014] [Accepted: 08/06/2014] [Indexed: 12/15/2022]
Abstract
Current smoking cessation pharmacotherapies have modest efficacy, and most smokers relapse within the first few days after a quit attempt. Nicotine withdrawal-induced craving and cognitive impairments predict smoking relapse during abstinence and suggest that cognitive-enhancing drugs may prevent relapse. ABT-089 and ABT-107 are subtype-selective nAChR agonists that improve cognitive performance in laboratory animals. However, there are no studies examining the effects of ABT-089 and ABT-107 on nicotine self-administration and the reinstatement of nicotine-seeking behavior, an animal model of relapse in human smokers. The goal of the present study was to determine the effects of the α4β2*/α6β2* nAChR agonist ABT-089 and the α7 nAChR agonist ABT-107 on nicotine taking and seeking in rats. The effects of acute ABT-089 and ABT-107 pretreatment on nicotine self-administration and reinstatement were tested in male Sprague Dawley rats. Parallel studies of ABT-089 and ABT-107 on sucrose self-administration and reinstatement were tested in separate groups of rats to determine if the effects of these drug treatments generalized to other reinforced behaviors. Nicotine and sucrose self-administration behaviors were not altered following acute administration of ABT-089 (0, 0.12, 1.2 and 12.0mg/kg) or ABT-107 (0, 0.03 and 0.3mg/kg). In contrast, both ABT-089 and ABT-107 pretreatment dose-dependently attenuated nicotine reinstatement. These effects were reinforcer-specific as no effects of ABT-089 or ABT-107 pretreatment on sucrose seeking were noted. Taken together, these findings suggest that ABT-089 and ABT-107 do not affect nicotine consumption, but may reduce the likelihood that a smoking lapse will lead to relapse.
Collapse
Affiliation(s)
- Alycia M Lee
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 125 South 31st Street, TRL Building, Rm 2214, Philadelphia, PA 19104, United States
| | - Adrian C Arreola
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 125 South 31st Street, TRL Building, Rm 2214, Philadelphia, PA 19104, United States
| | - Blake A Kimmey
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 125 South 31st Street, TRL Building, Rm 2214, Philadelphia, PA 19104, United States
| | - Heath D Schmidt
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, 125 South 31st Street, TRL Building, Rm 2214, Philadelphia, PA 19104, United States.
| |
Collapse
|
45
|
Kimmey BA, Rupprecht LE, Hayes MR, Schmidt HD. Donepezil, an acetylcholinesterase inhibitor, attenuates nicotine self-administration and reinstatement of nicotine seeking in rats. Addict Biol 2014; 19:539-51. [PMID: 23231479 DOI: 10.1111/adb.12014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nicotine craving and cognitive impairments represent core symptoms of nicotine withdrawal and predict relapse in abstinent smokers. Current smoking cessation pharmacotherapies have limited efficacy in preventing relapse and maintaining abstinence during withdrawal. Donepezil is an acetylcholinesterase inhibitor that has been shown previously to improve cognition in healthy non-treatment-seeking smokers. However, there are no studies examining the effects of donepezil on nicotine self-administration and/or the reinstatement of nicotine-seeking behavior in rodents. The present experiments were designed to determine the effects of acute donepezil administration on nicotine taking and the reinstatement of nicotine-seeking behavior, an animal model of relapse in abstinent human smokers. Moreover, the effects of acute donepezil administration on sucrose self-administration and sucrose seeking were also investigated in order to determine whether donepezil's effects generalized to other reinforced behaviors. Acute donepezil administration (1.0 or 3.0 mg/kg, i.p.) attenuated nicotine, but not sucrose self-administration maintained on a fixed-ratio 5 schedule of reinforcement. Donepezil administration also dose-dependently attenuated the reinstatement of both nicotine- and sucrose-seeking behaviors. Commonly reported adverse effects of donepezil treatment in humans are nausea and vomiting. However, at doses required to attenuate nicotine self-administration in rodents, no effects of donepezil on nausea/malaise as measured by pica were observed. Collectively, these results indicate that increased extracellular acetylcholine levels are sufficient to attenuate nicotine taking and seeking in rats and that these effects are not due to adverse malaise symptoms such as nausea.
Collapse
Affiliation(s)
- Blake A. Kimmey
- Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Laura E. Rupprecht
- Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Matthew R. Hayes
- Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Heath D. Schmidt
- Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| |
Collapse
|
46
|
Mak S, Luk WWK, Cui W, Hu S, Tsim KWK, Han Y. Synergistic Inhibition on Acetylcholinesterase by the Combination of Berberine and Palmatine Originally Isolated from Chinese Medicinal Herbs. J Mol Neurosci 2014; 53:511-6. [DOI: 10.1007/s12031-014-0288-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/13/2014] [Indexed: 11/29/2022]
|
47
|
Ashare RL, Schmidt HD. Optimizing treatments for nicotine dependence by increasing cognitive performance during withdrawal. Expert Opin Drug Discov 2014; 9:579-94. [PMID: 24707983 DOI: 10.1517/17460441.2014.908180] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, there is a clear need to develop novel antismoking medications. Nicotine withdrawal is associated with cognitive impairments that predict smoking relapse. It has been proposed that these cognitive deficits are a hallmark of nicotine withdrawal that could be targeted in order to prevent smoking relapse. Thus, pharmacotherapies that increase cognitive performance during nicotine withdrawal may represent potential smoking cessation agents. AREAS COVERED The authors review the clinical literature demonstrating that nicotine withdrawal is associated with deficits in working memory, attention and response inhibition. They then briefly summarize different classes of compounds and strategies to increase cognitive performance during nicotine withdrawal. Particular emphasis has been placed on translational research in order to highlight areas for which there is strong rationale for pilot clinical trials of potential smoking cessation medications. EXPERT OPINION There is emerging evidence that supports deficits in cognitive function as a plausible nicotine withdrawal phenotype. The authors furthermore believe that the translational paradigms presented here may represent efficient and valid means for the evaluation of cognitive-enhancing medications as possible treatments for nicotine dependence.
Collapse
Affiliation(s)
- Rebecca L Ashare
- University of Pennsylvania, Perelman School of Medicine, Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry , 3535 Market St, Suite 4100, Philadelphia, PA 19104 , USA +1 215 746 5789 ;
| | | |
Collapse
|
48
|
Fowler CD, Kenny PJ. Nicotine aversion: Neurobiological mechanisms and relevance to tobacco dependence vulnerability. Neuropharmacology 2014; 76 Pt B:533-44. [PMID: 24055497 PMCID: PMC3858456 DOI: 10.1016/j.neuropharm.2013.09.008] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/04/2013] [Accepted: 09/05/2013] [Indexed: 11/22/2022]
Abstract
Nicotine stimulates brain reward circuitries, most prominently the mesocorticolimbic dopamine system, and this action plays a critical in establishing and maintaining the tobacco smoking habit. Compounds that attenuate nicotine reward are considered promising therapeutic candidates for tobacco dependence, but many of these agents have other actions that limit their potential utility. Nicotine is also highly noxious, particularly at higher doses, and aversive reactions to nicotine after initial exposure can decrease the likelihood of developing a tobacco habit in many first time smokers. Nevertheless, relatively little is known about the mechanisms of nicotine aversion. The purpose of this review is to present recent new insights into the neurobiological mechanisms that regulate avoidance of nicotine. First, the role of the mesocorticolimbic system, so often associated with nicotine reward, in regulating nicotine aversion is highlighted. Second, genetic variation that modifies noxious responses to nicotine and thereby influences vulnerability to tobacco dependence, in particular variation in the CHRNA5-CHRNA3-CHRNB4 nicotinic acetylcholine receptor (nAChR) subunit gene cluster, will be discussed. Third, the role of the habenular complex in nicotine aversion, primarily medial habenular projections to the interpeduncular nucleus (IPN) but also lateral habenular projections to rostromedial tegmental nucleus (RMTg) and ventral tegmental area (VTA) are reviewed. Forth, brain circuits that are enriched in nAChRs, but whose role in nicotine avoidance has not yet been assessed, will be identified. Finally, the feasibility of developing novel therapeutic agents for tobacco dependence that act not by blocking nicotine reward but by enhancing nicotine avoidance will be considered. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Christie D Fowler
- Laboratory of Behavioral and Molecular Neuroscience, Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | | |
Collapse
|
49
|
Liu X. Positive allosteric modulation of α4β2 nicotinic acetylcholine receptors as a new approach to smoking reduction: evidence from a rat model of nicotine self-administration. Psychopharmacology (Berl) 2013; 230:203-13. [PMID: 23712602 PMCID: PMC3797181 DOI: 10.1007/s00213-013-3145-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 05/06/2013] [Indexed: 12/14/2022]
Abstract
RATIONALE The α4β2 subtype of nicotinic acetylcholine receptors (nAChRs) plays a central role in the mediation of nicotine reinforcement. Positive allosteric modulators (PAMs) at α4β2 nAChRs facilitate the intrinsic efficiency of these receptors, although they do not directly activate the receptors. α4β2 PAMs are hypothesized to reduce nicotine self-administration in subjects engaged in routine nicotine consumption. The present study tested this hypothesis using a rat model of nicotine self-administration. METHODS Male Sprague-Dawley rats were trained in daily 1-h sessions to intravenously self-administer nicotine (0.03 mg/kg per infusion, free base) on a fixed-ratio 5 schedule. The effects of the α4β2 PAM desformylflustrabromine (dFBr), α4β2 agonist 5-iodo-A-85380, and acetylcholinesterase inhibitor galantamine on nicotine intake were examined. The ability of dFBr and 5-iodo-A-85380 to substitute for nicotine was also assessed. RESULTS dFBr and 5-iodo-A-85380 dose-dependently reduced nicotine self-administration without changing lever responses for food. Galantamine decreased the self-administration of nicotine and food at high doses. Unlike 5-iodo-A-85380, dFBr failed to substitute for nicotine in supporting self-administration behavior. CONCLUSIONS These results demonstrated the effectiveness of dFBr in reducing nicotine intake and the inability of dFBr to support self-administration behavior. These findings suggest that positive allosteric modulation of α4β2 nAChRs may be a promising target for the treatment of nicotine addiction. Moreover, α4β2 PAMs, in contrast to agonist medications, may have clinical advantages because they may have little liability for abuse because of their lack of reinforcing actions on their own.
Collapse
Affiliation(s)
- Xiu Liu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA,
| |
Collapse
|
50
|
Perez XA, McIntosh JM, Quik M. Long-term nicotine treatment down-regulates α6β2* nicotinic receptor expression and function in nucleus accumbens. J Neurochem 2013; 127:762-71. [PMID: 23992036 DOI: 10.1111/jnc.12442] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/26/2013] [Accepted: 08/28/2013] [Indexed: 12/31/2022]
Abstract
Long-term nicotine exposure induces alterations in dopamine transmission in nucleus accumbens that sustain the reinforcing effects of smoking. One approach to understand the adaptive changes that arise involves measurement of endogenous dopamine release using voltammetry. We therefore treated rats for 2-3 months with nicotine and examined alterations in nAChR subtype expression and electrically evoked dopamine release in rat nucleus accumbens shell, a region key in addiction. Long-term nicotine treatment selectively decreased stimulated α6β2* nAChR-mediated dopamine release compared with vehicle-treated rats. It also reduced α6β2* nAChRs, suggesting the receptor decline may contribute to the functional loss. This decreased response in release after chronic nicotine treatment was still partially sensitive to the agonist nicotine. Studies with an acetylcholinesterase inhibitor demonstrated that the response was also sensitive to increased endogenous acetylcholine. However, unlike the agonists, nAChR antagonists decreased dopamine release only in vehicle- but not nicotine-treated rats. As antagonists function by blocking the action of acetylcholine, their ineffectiveness suggests that reduced acetylcholine levels partly underlie the dampened α6β2* nAChR-mediated function in nicotine-treated rats. As long-term nicotine modifies dopamine release by decreasing α6β2* nAChRs and their function, these data suggest that interventions that target this subtype may be useful for treating nicotine dependence. Long-term nicotine treatment decreases dopamine (DA) transmission in the mesolimbic dopaminergic system. Our data suggest this may involve a decrease in α6β2* nicotinic receptor expression and function. These changes may play a key role in nicotine reward and dependence.
Collapse
Affiliation(s)
- Xiomara A Perez
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | | | | |
Collapse
|