1
|
Nourirad SN, Bayat AH, Sani M, Beirami A, Hasanzadeh M, Roustaee S, Fathi M, Vakili K, Parvardeh S, Meftahi GH, Sarahian N, Khodagholi F, Aliaghaei A, Abbaszadeh HA, Moghaddam MH. Memory loss induced by lisdexamfetamine in the rat: A behavioral, electrophysiological, and histopathological Study. Behav Brain Res 2024; 465:114963. [PMID: 38499158 DOI: 10.1016/j.bbr.2024.114963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Lisdexamfetamine (LDX) is one of the drugs commonly used to treat attention deficit hyperactivity disorder (ADHD). However, its neurological side effects, particularly on cognition, are not fully understood. The present study focused on memory in rats treated with four weeks of LDX injection. We compared LDX-treated rats with control ones, using several methods to evaluate the behavioral responses and electrophysiological, molecular, and histological properties in the hippocampus. Our findings demonstrated that subchronic administration of LDX impaired behavioral performance in all memory assessment tests (Y maze, Morris Water Maze, and Shuttle box). Although LDX did not alter population spike (PS) amplitude, it increased the field excitatory postsynaptic potential (fEPSP) slope of evoked potentials of LTP components. Also, in addition to an increase in expression of caspase-3 in the hippocampus, which indicates the susceptibility to apoptosis in LDX-treated rats, the number of microglia and astrocytes went up significantly in the LDX group. Moreover, Sholl's analysis showed an increase in the soma size and total process length in both hippocampal astrocytes and microglia. Overall, because of these destructive effects of LDX on the hippocampus, which is one of the critical memory-related areas of the brain, the findings of this investigation provide evidence to show the disruption of memory-related variables following the LDX. However, more research is needed to clarify it.
Collapse
Affiliation(s)
- Seyedeh Naghmeh Nourirad
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Basic Sciences, Saveh University of Medical Sciences, Saveh, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Amirreza Beirami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maral Hasanzadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Roustaee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nahid Sarahian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hojjat-Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Blasco-Fontecilla H. Is Histamine and Not Acetylcholine the Missing Link between ADHD and Allergies? Speer Allergic Tension Fatigue Syndrome Re-Visited. J Clin Med 2023; 12:5350. [PMID: 37629392 PMCID: PMC10455974 DOI: 10.3390/jcm12165350] [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: 05/28/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Speer allergic tension-fatigue syndrome (SATFS) is a classic allergy syndrome characterized by allergy-like symptoms, muscle tension, headaches, chronic fatigue, and other particular behaviors that were initially described in the fifties. The particular behaviors displayed include symptoms such as hyperkinesis, hyperesthesia (i.e., insomnia), restlessness, and distractibility, among others. Interestingly, these symptoms are very similar to descriptions of attention deficit hyperactivity disorder (ADHD), the most prevalent neurodevelopmental disorder worldwide, which is characterized by inattention, hyperactivity, and impulsivity. The clinical description of SATFS precedes the nomination of ADHD in 1960 by Stella Chess. In this conceptual paper, we stress that there is a gap in the research on the relationship between ADHD and allergic pathologies. The hypotheses of this conceptual paper are (1) SATFS is probably one of the first and best historical descriptions of ADHD alongside a common comorbidity (allergy) displayed by these patients; (2) SATFS (ADHD) is a systemic disease that includes both somatic and behavioral manifestations that may influence each other in a bidirectional manner; (3) The role of neuroinflammation and histamine is key for understanding the pathophysiology of ADHD and its frequent somatic comorbidities; (4) The deficiency of the diamine oxidase (DAO) enzyme, which metabolizes histamine extracellularly, may play a role in the pathophysiology of ADHD. Decreased DAO activity may lead to an accumulation of histamine, which could contribute to core ADHD symptoms and comorbid disorders. Further empirical studies are needed to confirm our hypotheses.
Collapse
Affiliation(s)
- Hilario Blasco-Fontecilla
- Department of Psychiatry, School of Medicine, Autonoma University of Madrid, 28049 Madrid, Spain; ; Tel.: +34-911916012
- Department of Psychiatry, Puerta de Hierro University Hospital, Health Research Institute Puerta de Hierro-Segovia de Arana (IDIPHISA), Majadahonda, 28222 Madrid, Spain
- ITA Mental Health, Korian, 28043 Madrid, Spain
- Center of Biomedical Network Research on Mental Health (CIBERSAM), 28029 Madrid, Spain
| |
Collapse
|
3
|
Gutiérrez-Casares JR, Quintero J, Segú-Vergés C, Rodríguez Monterde P, Pozo-Rubio T, Coma M, Montoto C. In silico clinical trial evaluating lisdexamfetamine's and methylphenidate's mechanism of action computational models in an attention-deficit/hyperactivity disorder virtual patients' population. Front Psychiatry 2023; 14:939650. [PMID: 37333910 PMCID: PMC10273406 DOI: 10.3389/fpsyt.2023.939650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/21/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is an impairing psychiatric condition with the stimulants, lisdexamfetamine (LDX), and methylphenidate (MPH), as the first lines pharmacological treatment. Methods Herein, we applied a novel in silico method to evaluate virtual LDX (vLDX) and vMPH as treatments for ADHD applying quantitative systems pharmacology (QSP) models. The objectives were to evaluate the model's output, considering the model characteristics and the information used to build them, to compare both virtual drugs' efficacy mechanisms, and to assess how demographic (age, body mass index, and sex) and clinical characteristics may affect vLDX's and vMPH's relative efficacies. Results and Discussion We molecularly characterized the drugs and pathologies based on a bibliographic search, and generated virtual populations of adults and children-adolescents totaling 2,600 individuals. For each virtual patient and virtual drug, we created physiologically based pharmacokinetic and QSP models applying the systems biology-based Therapeutic Performance Mapping System technology. The resulting models' predicted protein activity indicated that both virtual drugs modulated ADHD through similar mechanisms, albeit with some differences. vMPH induced several general synaptic, neurotransmitter, and nerve impulse-related processes, whereas vLDX seemed to modulate neural processes more specific to ADHD, such as GABAergic inhibitory synapses and regulation of the reward system. While both drugs' models were linked to an effect over neuroinflammation and altered neural viability, vLDX had a significant impact on neurotransmitter imbalance and vMPH on circadian system deregulation. Among demographic characteristics, age and body mass index affected the efficacy of both virtual treatments, although the effect was more marked for vLDX. Regarding comorbidities, only depression negatively impacted both virtual drugs' efficacy mechanisms and, while that of vLDX were more affected by the co-treatment of tic disorders, the efficacy mechanisms of vMPH were disturbed by wide-spectrum psychiatric drugs. Our in silico results suggested that both drugs could have similar efficacy mechanisms as ADHD treatment in adult and pediatric populations and allowed raising hypotheses for their differential impact in specific patient groups, although these results require prospective validation for clinical translatability.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Universidad Complutense, Madrid, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Medical Department, Takeda Farmacéutica España, Madrid, Spain
| |
Collapse
|
4
|
Zhang DP, Lu XY, He SC, Li WY, Ao R, Leung FCY, Zhang ZM, Chen QB, Zhang SJ. Sodium tanshinone IIA sulfonate protects against Aβ-induced cell toxicity through regulating Aβ process. J Cell Mol Med 2020; 24:3328-3335. [PMID: 31989795 PMCID: PMC7131914 DOI: 10.1111/jcmm.15006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/09/2019] [Accepted: 01/06/2020] [Indexed: 01/14/2023] Open
Abstract
Sodium tanshinone IIA sulfonate (STS) has been reported to prevent Alzheimer's disease (AD). However, the mechanism is still unknown. In this study, two in vitro models, Aβ-treated SH-SY5Y cells and SH-SY5Y human neuroblastoma cells transfected with APPsw (SH-SY5Y-APPsw cells), were employed to investigate the neuroprotective of STS. The results revealed that pretreatment with STS (1, 10 and 100 µmol/L) for 24 hours could protect against Aβ (10 µmol/L)-induced cell toxicity in a dose-dependent manner in the SH-SY5Y cells. Sodium tanshinone IIA sulfonate decreased the concentrations of reactive oxygen species, malondialdehyde, NO and iNOS, while increased the activities of superoxide dismutase and glutathione peroxidase in the SH-SY5Y cells. Sodium tanshinone IIA sulfonate decreased the levels of inflammatory factors (IL-1β, IL-6 and TNF-α) in the SH-SY5Y cells. In addition, Western blot results revealed that the expressions of neprilysin and insulin-degrading enzyme were up-regulated in the SH-SY5Y cells after STS treatment. Furthermore, ELISA and Western blot results showed that STS could decrease the levels of Aβ. ELISA and qPCR results indicated that STS could increase α-secretase (ADAM10) activity and decrease β-secretase (BACE1) activity. In conclusion, STS could protect against Aβ-induced cell damage by modulating Aβ degration and generation. Sodium tanshinone IIA sulfonate could be a promising candidate for AD treatment.
Collapse
Affiliation(s)
- Da-Peng Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin-Yi Lu
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Chen He
- Department of Neurology, The People's Hospital of Baiyun District Guangzhou, Guangzhou, China
| | - Wan-Yan Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ran Ao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feona Chung-Yin Leung
- LKS Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhi-Min Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qu-Bo Chen
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shi-Jie Zhang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Development of a non-human primate model to support CNS translational research: Demonstration with D-amphetamine exposure and dopamine response. J Neurosci Methods 2019; 317:71-81. [PMID: 30768951 DOI: 10.1016/j.jneumeth.2019.02.005] [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: 02/06/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND Challenges specific to the discovery and development of candidate CNS drugs have led to implementation of various in silico, in vitro and in vivo approaches to improve the odds for commercialization of novel treatments. NEW METHOD Advances in analytical methodology and microdialysis probe design have enabled development of a non-human primate model capable of measuring concentrations of drugs or endogenous chemicals in brain extracellular fluid (ECF) and cerebrospinal fluid (CSF). Linking these to population modeling reduces animal numbers to support predictive translational sciences in primates. Application to measure D-amphetamine exposure and dopamine response in ECF and CSF demonstrate the approach. RESULTS Following a 0.1 mg/kg intravenous bolus dose of D-amphetamine, a population approach was used to build a plasma compartmental-based and brain physiologic-based pharmacokinetic (PK) model linking drug concentrations in plasma to brain ECF and CSF concentrations. Dopamine was also measured in brain ECF. The PK model was used to simulate the relationship between D-amphetamine exposure and dopamine response in ECF over a wide dose range. COMPARISONS WITH EXISTING METHODS Ability to co-sample and measure drug and endogenous substances in blood, brain ECF and/or CSF, coupled with population modeling, provides an in vivo approach to evaluate CNS drug penetration and effect in non-human primates. CONCLUSIONS A method to measure drug and endogenous neurochemicals in non-human primate brain fluids is demonstrated. Its basis in non-human primates merits improved confidence regarding predictions of drug exposure and target engagement in human CNS.
Collapse
|
6
|
CGY-1, a biflavonoid isolated from cardiocrinum giganteum seeds, improves memory deficits by modulating the cholinergic system in scopolamine-treated mice. Biomed Pharmacother 2019; 111:496-502. [DOI: 10.1016/j.biopha.2018.12.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/19/2018] [Accepted: 12/23/2018] [Indexed: 12/26/2022] Open
|
7
|
van Gaalen MM, Schlumbohm C, Folgering JH, Adhikari S, Bhattacharya C, Steinbach D, Stratford RE. Development of a Semimechanistic Pharmacokinetic-Pharmacodynamic Model Describing Dextroamphetamine Exposure and Striatal Dopamine Response in Rats and Nonhuman Primates following a Single Dose of Dextroamphetamine. J Pharmacol Exp Ther 2019; 369:107-120. [PMID: 30733244 DOI: 10.1124/jpet.118.254508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/23/2019] [Indexed: 11/22/2022] Open
Abstract
Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. A semimechanistic pharmacokinetic and pharmacodynamic (PK/PD) model with consideration of these multiple effects as a basis was developed. Integrated pharmacokinetics of d-amphetamine in plasma, brain extracellular fluid (ECF) via microdialysis, and cerebrospinal fluid were characterized using a population approach. This PK model was then linked to an indirect-response pharmacodynamic model using as a basis the measurement of extracellular striatal dopamine, also via microdialysis. In both rats and nonhuman primates (NHPs), d-amphetamine stimulation of dopamine outflow (reverse transport) through DAT was primarily responsible for the dose-linear increase in dopamine. As well, in both species a moderator function was needed to account for loss of the dopamine response in the presence of a relatively sustained d-amphetamine ECF exposure, presumptive of an acute tolerance response. PK/PD model structure was consistent between species; however, there was a 10-fold faster return to baseline dopamine in NHPs in response to an acute d-amphetamine challenge. These results suggest preservation from rodents to NHPs regarding the mechanism by which amphetamine increases extracellular dopamine, but a faster system response in NHPs to tolerate this increase. This microdialysis-based PK/PD model suggests greater value in directing preclinical discovery of novel approaches that modify reverse transport stimulation to treat amphetamine abuse. General value regarding insertion of an NHP model in paradigm rodent-to-human translational research is also suggested.
Collapse
Affiliation(s)
- Marcel M van Gaalen
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Christina Schlumbohm
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Joost H Folgering
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Saugat Adhikari
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Chandrali Bhattacharya
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Douglas Steinbach
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Robert E Stratford
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| |
Collapse
|
8
|
Willemze RA, Welting O, van Hamersveld HP, Meijer SL, Folgering JHA, Darwinkel H, Witherington J, Sridhar A, Vervoordeldonk MJ, Seppen J, de Jonge WJ. Neuronal control of experimental colitis occurs via sympathetic intestinal innervation. Neurogastroenterol Motil 2018; 30. [PMID: 28745812 DOI: 10.1111/nmo.13163] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/20/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Vagus nerve stimulation is currently clinically evaluated as a treatment for inflammatory bowel disease. However, the mechanism by which this therapeutic intervention can have an immune-regulatory effect in colitis remains unclear. We determined the effect of intestine-specific vagotomy or intestine-specific sympathectomy of the superior mesenteric nerve (SMN) on dextran sodium sulfate (DSS)-induced colitis in mice. Furthermore, we tested the efficacy of therapeutic SMN stimulation to treat DSS-induced colitis in rats. METHODS Vagal and SMN fibers were surgically dissected to achieve intestine-specific vagotomy and sympathectomy. Chronic SMN stimulation was achieved by implantation of a cuff electrode. Stimulation was done twice daily for 5 minutes using a biphasic pulse (10 Hz, 200 μA, 2 ms). Disease activity index (DAI) was used as a clinical parameter for colitis severity. Colonic cytokine expression was measured by quantitative PCR and ELISA. KEY RESULTS Intestine-specific vagotomy had no effect on DSS-induced colitis in mice. However, SMN sympathectomy caused a significantly higher DAI compared to sham-operated mice. Conversely, SMN stimulation led to a significantly improved DAI compared to sham stimulation, although no other parameters of colitis were affected significantly. CONCLUSIONS & INFERENCES Our results indicate that sympathetic innervation regulates the intestinal immune system as SMN denervation augments, and SMN stimulation ameliorates DSS-induced colitis. Surprisingly, intestine-specific vagal nerve denervation had no effect in DSS-induced colitis.
Collapse
Affiliation(s)
- R A Willemze
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - O Welting
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - H P van Hamersveld
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - S L Meijer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - H Darwinkel
- Brains On-Line B.V., Groningen, The Netherlands
| | | | - A Sridhar
- Galvani Bioelectronics, Stevenage, UK
| | | | - J Seppen
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Abstract
Binge-eating disorder (BED) is the most prevalent eating disorder with estimates of 2-5% of the general adult population. Nonetheless, its pathophysiology is poorly understood. Furthermore, there exist few therapeutic options for its effective treatment. Here we review the current state of binge-eating neurobiology and pharmacology, drawing from clinical therapeutic, neuroimaging, cognitive, human genetic and animal model studies. These studies, which are still in their infancy, indicate that while there are many gaps in our knowledge, several key neural substrates appear to underpin binge-eating and may be conserved between human and animals. This observation suggests that behavioral intermediate phenotypes or endophenotypes relevant to BED may be modeled in animals, facilitating the identification and testing of novel pharmacological targets. The development of novel, safe and effective pharmacological therapies for the treatment of BED will enhance the ability of clinicians to provide optimal care for people with BED.
Collapse
Affiliation(s)
- Peter H Hutson
- Department of Neurobiology, CNS Discovery, Teva Pharmaceuticals, West Chester, PA, USA.
| | - Iris M Balodis
- Peter Boris Centre for Addiction Research, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Marc N Potenza
- Department of Psychiatry, Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; National Center on Addiction and Substance Abuse, USA; Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
10
|
Hutson PH, Rowley HL, Gosden J, Kulkarni RS, Slater N, Love PL, Wang Y, Heal D. The effects in rats of lisdexamfetamine in combination with olanzapine on mesocorticolimbic dopamine efflux, striatal dopamine D2 receptor occupancy and stimulus generalization to a D-amphetamine cue. Neuropharmacology 2015; 101:24-35. [PMID: 26384654 DOI: 10.1016/j.neuropharm.2015.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/28/2015] [Accepted: 09/11/2015] [Indexed: 12/21/2022]
Abstract
The etiology of schizophrenia is poorly understood and two principle hypotheses have dominated the field. Firstly, that subcortical dopamine function is enhanced while cortical dopamine function is reduced and secondly, that cortical glutamate systems are dysfunctional. It is also widely accepted that currently used antipsychotics have essentially no impact on cognitive deficits and persistent negative symptoms in schizophrenia. Reduced dopamine transmission via dopamine D1 receptors in the prefrontal cortex has been hypothesized to be involved in the aetiology of these symptom domains and enhancing cortical dopamine transmission within an optimal window has been suggested to be potentially beneficial. In these pre-clinical studies we have determined that combined administration of the d-amphetamine pro-drug, lisdexamfetamine and the atypical antipsychotic olanzapine increased dopamine efflux in the rat prefrontal cortex and nucleus accumbens to an extent greater than either drug given separately without affecting olanzapine's ability to block striatal dopamine D2 receptors which is important for its antipsychotic activity. Furthermore, in an established rodent model used to compare the subjective effects of novel compounds the ability of lisdexamfetamine to generalize to a d-amphetamine cue was dose-dependently attenuated when co-administered with olanzapine suggesting that lisdexamfetamine may produce less marked subjective effects when administered adjunctively with olanzapine.
Collapse
Affiliation(s)
- Peter H Hutson
- Shire Pharmaceutical, 300 Shire Way, Lexington, MA 02421 USA.
| | - Helen L Rowley
- RenaSci Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham, NG1 1GF, UK
| | - James Gosden
- RenaSci Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham, NG1 1GF, UK
| | - Rajiv S Kulkarni
- RenaSci Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham, NG1 1GF, UK
| | - Nigel Slater
- RenaSci Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham, NG1 1GF, UK
| | - Patrick L Love
- Covance, Inc., 671 South Meridian Road, Greenfield, IN 46140, USA
| | - Yiyun Wang
- Covance, Inc., 671 South Meridian Road, Greenfield, IN 46140, USA
| | - David Heal
- RenaSci Ltd, BioCity, Nottingham, Pennyfoot Street, Nottingham, NG1 1GF, UK
| |
Collapse
|