1
|
Hersey M, Tanda G. Modafinil, an atypical CNS stimulant? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:287-326. [PMID: 38467484 DOI: 10.1016/bs.apha.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Modafinil is a central nervous system stimulant approved for the treatment of narcolepsy and sleep disorders. Due to its wide range of biochemical actions, modafinil has been explored for other potential therapeutic uses. Indeed, it has shown promise as a therapy for cognitive disfunction resulting from neurologic disorders like ADHD, and as a smart drug in non-medical settings. The mechanism(s) of actions underlying the therapeutic efficacy of this agent remains largely elusive. Modafinil is known to inhibit the dopamine transporter, thus decreasing dopamine reuptake following neuronal release, an effect shared by addictive psychostimulants. However, modafinil is unique in that only a few cases of dependence on this drug have been reported, as compared to other psychostimulants. Moreover, modafinil has been tested, with some success, as a potential therapeutic agent to combat psychostimulant and other substance use disorders. Modafinil has additional, but less understood, actions on other neurotransmitter systems (GABA, glutamate, serotonin, norepinephrine, etc.). These interactions, together with its ability to activate selected brain regions, are likely one of the keys to understand its unique pharmacology and therapeutic activity as a CNS stimulant. In this chapter, we outline the pharmacokinetics and pharmacodynamics of modafinil that suggest it has an "atypical" CNS stimulant profile. We also highlight the current approved and off label uses of modafinil, including its beneficial effects as a treatment for sleep disorders, cognitive functions, and substance use disorders.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, NIDA-IRP, NIH, Baltimore, MD, United States.
| |
Collapse
|
2
|
Martins D, Brodmann K, Veronese M, Dipasquale O, Mazibuko N, Schuschnig U, Zelaya F, Fotopoulou A, Paloyelis Y. "Less is more": a dose-response account of intranasal oxytocin pharmacodynamics in the human brain. Prog Neurobiol 2022; 211:102239. [PMID: 35122880 DOI: 10.1016/j.pneurobio.2022.102239] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/23/2022] [Accepted: 01/31/2022] [Indexed: 12/27/2022]
Abstract
Intranasal oxytocin is attracting attention as a potential treatment for several brain disorders due to promising preclinical results. However, translating findings to humans has been hampered by remaining uncertainties about its pharmacodynamics and the methods used to probe its effects in the human brain. Using a dose-response design (9, 18 and 36 IU), we demonstrate that intranasal oxytocin-induced changes in local regional cerebral blood flow (rCBF) in the amygdala at rest, and in the covariance between rCBF in the amygdala and other key hubs of the brain oxytocin system, follow a dose-response curve with maximal effects for lower doses. Yet, the effects on local rCBF might vary by amygdala subdivision, highlighting the need to qualify dose-response curves within subregion. We further link physiological changes with the density of the oxytocin receptor gene mRNA across brain regions, strengthening our confidence in intranasal oxytocin as a valid approach to engage central targets. Finally, we demonstrate that intranasal oxytocin does not disrupt cerebrovascular reactivity, which corroborates the validity of haemodynamic neuroimaging to probe the effects of intranasal oxytocin in the human brain. DATA AVAILABILITY: Participants did not consent for open sharing of the data. Therefore, data can only be accessed from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Katja Brodmann
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ndaba Mazibuko
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | | | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Aikaterini Fotopoulou
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Yannis Paloyelis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.
| |
Collapse
|
3
|
Cruces-Solis H, Nissen W, Ferger B, Arban R. Whole-brain signatures of functional connectivity after bidirectional modulation of the dopaminergic system in mice. Neuropharmacology 2020; 178:108246. [PMID: 32771528 DOI: 10.1016/j.neuropharm.2020.108246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
While neuropsychiatric drugs influence neural activity across multiple brain regions, the current understanding of their mechanism of action derives from studies that investigate an influence of a given drug onto a pre-selected and small number of brain regions. To understand how neuropsychiatric drugs affect coordinated activity across brain regions and to detect the brain regions most relevant to pharmacological action in an unbiased way, studies that assess brain-wide neuronal activity are paramount. Here, we used whole-brain immunostaining of the neuronal activity marker cFOS, and graph theory to generate brain-wide maps of neuronal activity upon pharmacological challenges. We generated brain-wide maps 2.5 h after treatment of the atypical dopamine transporter inhibitor modafinil (10, 30, and 100 mg/kg) or the vesicular monoamine transporter 2 inhibitor tetrabenazine (0.25, 0.5 and 1 mg/kg). Modafinil increased the number of cFOS positive neurons in a dose-dependent manner. Moreover, modafinil significantly reduced functional connectivity across the entire brain. Graph theory analysis revealed that modafinil decreased the node degree of cortical and subcortical regions at the three doses tested, followed by a reduction in global efficiency. Simultaneously, we identified highly interconnected hub regions that emerge exclusively upon modafinil treatment. These regions were the mediodorsal thalamus, periaqueductal gray, subiculum, and rhomboid nucleus. On the other hand, while tetrabenazine had mild effects on cFOS counts, it reduced functional connectivity across the entire brain, cortical node degree, and global efficiency. As hub regions, we identified the substantia innominata and ventral pallidum. Our results uncovered novel mechanisms of action at a brain-wide scale for modafinil and tetrabenazine. Our analytical approach offers a tool to characterize signatures of whole-brain functional connectivity for drug candidates and to identify potential undesired effects at a mesoscopic scale. Additionally, it offers a guide towards targeted experiments on newly identified hub regions.
Collapse
Affiliation(s)
- Hugo Cruces-Solis
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany.
| | - Wiebke Nissen
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| | - Boris Ferger
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| | - Roberto Arban
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany.
| |
Collapse
|
4
|
Sagheddu C, Pintori N, Kalaba P, Dragačević V, Piras G, Lubec J, Simola N, De Luca MA, Lubec G, Pistis M. Neurophysiological and Neurochemical Effects of the Putative Cognitive Enhancer ( S)-CE-123 on Mesocorticolimbic Dopamine System. Biomolecules 2020; 10:biom10050779. [PMID: 32443397 PMCID: PMC7277835 DOI: 10.3390/biom10050779] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Treatments for cognitive impairments associated with neuropsychiatric disorders, such as attention deficit hyperactivity disorder or narcolepsy, aim at modulating extracellular dopamine levels in the brain. CE-123 (5-((benzhydrylsulfinyl)methyl) thiazole) is a novel modafinil analog with improved specificity and efficacy for dopamine transporter inhibition that improves cognitive and motivational processes in experimental animals. We studied the neuropharmacological and behavioral effects of the S-enantiomer of CE-123 ((S)-CE-123) and R-modafinil in cognitive- and reward-related brain areas of adult male rats. In vivo single unit recordings in anesthetized animals showed that (S)-CE-123, but not R-modafinil, dose-dependently (1.25 to 10 mg/kg i.v.) reduced firing of pyramidal neurons in the infralimbic/prelimbic (IL/PrL) cortex. Neither compound the affected firing activity of ventral tegmental area dopamine cells. In freely moving animals, (S)-CE-123 (10 mg/kg i.p.) increased extracellular dopamine levels in the IL/PrL, with different patterns when compared to R-modafinil (10 mg/kg i.p.); in the nucleus accumbens shell, a low and transitory increase of dopamine was observed only after (S)-CE-123. Neither (S)-CE-123 nor R-modafinil initiated the emission of 50-kHz ultrasonic vocalizations, a behavioral marker of positive affect and drug-mediated reward. Our data support previous reports of the procognitive effects of (S)-CE-123, and show a minor impact on reward-related dopaminergic areas.
Collapse
Affiliation(s)
- Claudia Sagheddu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Nicholas Pintori
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Vladimir Dragačević
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Gessica Piras
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Jana Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Nicola Simola
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| | - Marco Pistis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
- Neuroscience Institute, National Research Council of Italy (CNR), Section of Cagliari, 09100 Cagliari, Italy
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| |
Collapse
|
5
|
Heyer-Osorno R, Juárez J. Modafinil reduces choice impulsivity while increasing motor activity in preadolescent rats treated prenatally with alcohol. Pharmacol Biochem Behav 2020; 194:172936. [PMID: 32360693 DOI: 10.1016/j.pbb.2020.172936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
Rats exposed prenatally to alcohol show a reduction in the spontaneous activity of dopaminergic neurons of the ventral tegmental area (VTA), as well as greater impulsive behavior and motor activity, behavioral alterations that have been related to dopaminergic dysfunction. Modafinil (MOD) is a dopamine (DA) reuptake blocker prescribed to treat sleep disorders; however, in recent years it has been used for the treatment of ADHD with positive results. Also, studies in humans and rodents show beneficial effects on learning and attention; however, studies evaluating MOD effects on impulsivity are few and show contradictory results. The purpose of this work was to evaluate the effect of a daily dose of MOD (60 mg/kg i.g.) on cognitive (or choice) impulsivity and motor activity in male preadolescent rats exposed prenatally to alcohol or sucrose (isocaloric control). MOD reduced the impulsive responses in a delay discounting task (DDT) at the same time that increased the motor activity, in both healthy and prenatal alcohol treated rats; however, MOD reduced the response latency in DDT only in prenatal alcohol treated rats. This differential effect of DA activation on impulsivity and motor activity show that the MOD dose that improves the impulse control, does not necessarily decrease motor activity, and suggests a possible differential neural mechanism underlying the expression of these behaviors. On the other hand, the changes in the response latency, only in prenatal alcohol treated groups, suggest that decision-making in animals with a dopaminergic dysfunction is more susceptible to be affected by MOD action.
Collapse
Affiliation(s)
- Rocio Heyer-Osorno
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jorge Juárez
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
6
|
Acute and Repeated Intranasal Oxytocin Differentially Modulate Brain-wide Functional Connectivity. Neuroscience 2020; 445:83-94. [PMID: 31917352 DOI: 10.1016/j.neuroscience.2019.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022]
Abstract
Central release of the neuropeptide oxytocin (OXT) modulates neural substrates involved in socio-affective behavior. This property has prompted research into the use of intranasal OXT administration as an adjunctive therapy for brain conditions characterized by social impairment, such as autism spectrum disorders (ASD). However, the neural circuitry and brain-wide functional networks recruited by intranasal OXT administration remain elusive. Moreover, little is known of the neuroadaptive cascade triggered by long-term administration of this peptide at the network level. To address these questions, we applied fMRI-based circuit mapping in adult mice upon acute and repeated (seven-day) intranasal dosing of OXT. We report that acute and chronic OXT administration elicit comparable fMRI activity as assessed with cerebral blood volume mapping, but entail largely different patterns of brain-wide functional connectivity. Specifically, acute OXT administration focally boosted connectivity within key limbic components of the rodent social brain, whereas repeated dosing led to a prominent and widespread increase in functional connectivity, involving a strong coupling between the amygdala and extended cortical territories. Importantly, this connectional reconfiguration was accompanied by a paradoxical reduction in social interaction and communication in wild-type mice. Our results identify the network substrates engaged by exogenous OXT administration, and show that repeated OXT dosing leads to a substantial reconfiguration of brain-wide connectivity, entailing an aberrant functional coupling between cortico-limbic structures involved in socio-communicative and affective functions. Such divergent patterns of network connectivity might contribute to discrepant clinical findings involving acute or long-term OXT dosing in clinical populations.
Collapse
|
7
|
Ortiz‐Orendain J, Covarrubias‐Castillo SA, Vazquez‐Alvarez AO, Castiello‐de Obeso S, Arias Quiñones GE, Seegers M, Colunga‐Lozano LE. Modafinil for people with schizophrenia or related disorders. Cochrane Database Syst Rev 2019; 12:CD008661. [PMID: 31828767 PMCID: PMC6906203 DOI: 10.1002/14651858.cd008661.pub2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND People with schizophrenia have a range of different symptoms, including positive symptoms (hallucinations and delusions), negative symptoms (such as social withdrawal and lack of affect), and cognitive impairment. The standard medication for people with schizophrenia is antipsychotics. However, these medications may not be effective for all symptoms of schizophrenia, as cognitive and negative symptoms are usually hard to treat. Additional therapies or medications are available for the management of these symptoms. Modafinil, a wakefulness-promoting agent most frequently used in narcolepsy or shift work sleep disorder, is one intervention that is theorised to have an effect of these symptoms. OBJECTIVES The primary objective of this review was to assess the effects of modafinil for people with schizophrenia or related disorders. SEARCH METHODS On 27 April 2015, 24 May 2017, and 31 October 2019, we searched the Cochrane Schizophrenia Group's register of trials, which is based on regular searches of CENTRAL, MEDLINE, Embase, AMED, BIOSIS, CINAHL, PsycINFO, PubMed, and registries of clinical trials. There are no language, time, document type, or publication status limitations for the inclusion of records in the register. SELECTION CRITERIA We selected all randomised controlled trials comparing modafinil with placebo or other treatments for people with schizophrenia or schizophrenia-spectrum disorders. DATA COLLECTION AND ANALYSIS We independently extracted data from the included studies. We analysed dichotomous data using risk ratios (RR) and 95% confidence intervals (CI). We analysed continuous data using mean difference (MD) with a 95% CI. We used a random-effects model for the meta-analysis. We used GRADE to complete a 'Summary of findings' table and assessed risk of bias for the included studies. MAIN RESULTS Eleven studies including a total of 422 participants contributed to data analyses. Most studies had a small population size (average 38 people per study) and were of short duration. We also detected a high risk of bias for selective outcome reporting in just under 50% of the trials. We therefore rated the overall methodological quality of the included studies as low. We considered seven main outcomes of interest: clinically important change in overall mental state, clinically important change in cognitive functioning, incidence of a clinically important adverse effect/event, clinically important change in global state, leaving the study early for any reason, clinically important change in quality of life, and hospital admission. All studies assessed the effects of adding modafinil to participants' usual antipsychotic treatment compared to adding placebo to usual antipsychotic treatment. Six studies found that adding modafinil to antipsychotic treatment may have little or no effect on overall mental state of people with schizophrenia, specifically the risk of worsening psychosis (RR 0.91, 95% CI 0.28 to 2.98; participants = 209; studies = 6, low-quality evidence). Regarding the effect of modafinil on cognitive function, the trials did not report clinically important change data, but one study reported endpoint scores on the MATRICS Consensus Cognitive Battery (MCCB): in this study we found no clear difference in scores between modafinil and placebo treatment groups (MD -3.10, 95% CI -10.9 to 4.7; participants = 48; studies = 1, very low-quality evidence). Only one study (N = 35) reported adverse effect/event data. In this study one serious adverse event occurred in each group (RR 0.84, 95% CI 0.06 to 12.42; participants = 35; studies = 1, very low-quality evidence). One study measured change in global state using the Clinical Global Impression - Improvement Scale. This study found that adding modafinil to antipsychotic treatment may have little or no effect on global state (RR 6.36, 95% CI 0.94 to 43.07, participants = 21; studies = 1, very low-quality evidence). Nine studies found that modafinil has no effect on numbers of participants leaving the study early (RR 1.26, 95% CI 0.63 to 2.52 participants = 357; studies = 9, moderate-quality evidence). None of the trials reported clinically important change in quality of life, but one study did report quality of life using endpoint scores on the Quality of Life Inventory, finding no clear difference between treatment groups (MD -0.2, 95% CI -1.18 to 0.78; participants = 20; studies = 1, very low-quality evidence). Finally, one study reported data for number of participants needing hospitalisation: one participant in each group was hospitalised (RR 0.84, 95% CI 0.06 to 12.42; participants = 35; studies = 1, very low-quality evidence). AUTHORS' CONCLUSIONS Due to methodological issues, low sample size, and short duration of the clinical trials as well as high risk of bias for outcome reporting, most of the evidence available for this review is of very low or low quality. For results where quality is low or very low, we are uncertain or very uncertain if the effect estimates are true effects, limiting our conclusions. Specifically, we found that modafinil is no better or worse than placebo at preventing worsening of psychosis; however, we are uncertain about this result. We have more confidence that participants receiving modafinil are no more likely to leave a trial early than participants receiving placebo. However, we are very uncertain about the remaining equivocal results between modafinil and placebo for outcomes such as improvement in global state or cognitive function, incidence of adverse events, and changes in quality of life. More high-quality data are needed before firm conclusions regarding the effects of modafinil for people with schizophrenia or related disorders can be made.
Collapse
Affiliation(s)
- Javier Ortiz‐Orendain
- Mayo ClinicDepartment of Psychiatry and Psychology200 First Street SWRochesterMinnesotaUSA55905
| | - Sergio A. Covarrubias‐Castillo
- Hospital Civil de Guadalajara "Fray Antonio Alcalde"Department of PsychiatryHospital 278. El RetiroGuadalajaraJaliscoMexico44280
| | - Alan Omar Vazquez‐Alvarez
- Health Sciences University Center, University of GuadalajaraInstitute of Experimental and Clinical Therapeutics, Department of PhysiologyGuadalajaraJaliscoMexico
| | - Santiago Castiello‐de Obeso
- Western Institute of Technology and Higher Education (ITESO)Psychophysiology Laboratory#8585 Anillo Perif. Sur Manuel Gómez MorínCol. Santa María TequepexpanGuadalajaraJaliscoMexico45604
| | - Gustavo E Arias Quiñones
- University of GuadalajaraDepartment of Neurosurgery. Hospital Civil "Fray Antonio Alcalde"Hospital 278. El RetiroGuadalajaraJaliscoMexico44340
| | - Maya Seegers
- Ben Gurion UniversityMedical School for International HealthNew YorkUSA
| | - Luis Enrique Colunga‐Lozano
- McMaster UniversityDepartments of Health Research Methods, Evidence, and Impact1280 Main Street WestHamiltonOntarioCanadaL8S 4L8
| | | |
Collapse
|
8
|
Nakajima M, Schmitt LI, Feng G, Halassa MM. Combinatorial Targeting of Distributed Forebrain Networks Reverses Noise Hypersensitivity in a Model of Autism Spectrum Disorder. Neuron 2019; 104:488-500.e11. [PMID: 31648899 DOI: 10.1016/j.neuron.2019.09.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/03/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorder (ASD) is associated with noise hypersensitivity, the suboptimal extraction of meaningful signals in noisy environments. Because sensory filtering can involve distinct automatic and executive circuit mechanisms, however, developing circuit-specific therapeutic strategies for ASD noise hypersensitivity can be challenging. Here, we find that both of these processes are individually perturbed in one monogenic form of ASD, Ptchd1 deletion. Although Ptchd1 is preferentially expressed in the thalamic reticular nucleus during development, pharmacological rescue of thalamic perturbations in knockout (KO) mice only normalized automatic sensory filtering. By discovering a separate prefrontal perturbation in these animals and adopting a combinatorial pharmacological approach that also rescued its associated goal-directed noise filtering deficit, we achieved full normalization of noise hypersensitivity in this model. Overall, our work highlights the importance of identifying large-scale functional circuit architectures and utilizing them as access points for behavioral disease correction.
Collapse
Affiliation(s)
- Miho Nakajima
- McGovern Institute for Brain Research and the Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - L Ian Schmitt
- McGovern Institute for Brain Research and the Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research and the Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA; The Stanley Center for Psychiatric Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Michael M Halassa
- McGovern Institute for Brain Research and the Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA; The Stanley Center for Psychiatric Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
9
|
Bifone A, Gozzi A, Cippitelli A, Matzeu A, Domi E, Li H, Scuppa G, Cannella N, Ubaldi M, Weiss F, Ciccocioppo. phMRI, neurochemical and behavioral responses to psychostimulants distinguishing genetically selected alcohol-preferring from genetically heterogenous rats. Addict Biol 2019; 24:981-993. [PMID: 30328656 PMCID: PMC6697752 DOI: 10.1111/adb.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/27/2018] [Accepted: 07/23/2018] [Indexed: 01/01/2023]
Abstract
Alcoholism is often associated with other forms of drug abuse, suggesting that innate predisposing factors may confer vulnerability to addiction to diverse substances. However, the neurobiological bases of these factors remain unknown. Here, we have used a combination of imaging, neurochemistry and behavioral techniques to investigate responses to the psychostimulant amphetamine in Marchigian Sardinian (msP) alcohol-preferring rats, a model of vulnerability to alcoholism. Specifically, we employed pharmacological magnetic resonance imaging to investigate the neural circuits engaged by amphetamine challenge, and to relate functional reactivity to neurochemical and behavioral responses. Moreover, we studied self-administration of cocaine in the msP rats. We found stronger functional responses in the extended amygdala, alongside with increased release of dopamine in the nucleus accumbens shell and augmented vertical locomotor activity compared with controls. Wistar and msP rats did not differ in operant cocaine self-administration under short access (2 hours) conditions, but msP rats exhibited a higher propensity to escalate drug intake following long access (6 hours). Our findings suggest that neurobiological and genetic mechanisms that convey vulnerability to excessive alcohol drinking also facilitate the transition from psychostimulants use to abuse.
Collapse
Affiliation(s)
- A Bifone
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - A Gozzi
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - A Cippitelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - A Matzeu
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - E Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - H Li
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - G Scuppa
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - N Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - M Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, Camerino 62032, Italy
| | - F Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, United States
| | - Ciccocioppo
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| |
Collapse
|
10
|
González B, Torres OV, Jayanthi S, Gomez N, Sosa MH, Bernardi A, Urbano FJ, García-Rill E, Cadet JL, Bisagno V. The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:222-234. [PMID: 30056065 PMCID: PMC8424782 DOI: 10.1016/j.pnpbp.2018.07.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/02/2023]
Abstract
METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single-dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R-expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre-treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole-cell patch clamp in transgenic BAC-Drd1a-tdTomato mice showed that METH, but not modafinil, induced paired-pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects.
Collapse
Affiliation(s)
- Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, California, United States
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Máximo H Sosa
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Edgar García-Rill
- Center for Translational Neuroscience, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Verónica Bisagno
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Vodovar D, Duchêne A, Wimberley C, Leroy C, Pottier G, Dauvilliers Y, Giaume C, Lin JS, Mouthon F, Tournier N, Charvériat M. Cortico-Amygdala-Striatal Activation by Modafinil/Flecainide Combination. Int J Neuropsychopharmacol 2018; 21:687-696. [PMID: 29635319 PMCID: PMC6031015 DOI: 10.1093/ijnp/pyy027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 02/07/2023] Open
Abstract
Background Modafinil, a nonamphetaminic wake-promoting compound, is prescribed as first line therapy in narcolepsy, an invalidating disorder characterized by excessive daytime sleepiness and cataplexy. Although its mode of action remains incompletely known, recent studies indicated that modafinil modulates astroglial connexin-based gap junctional communication as administration of a low dose of flecainide, an astroglial connexin inhibitor, enhanced the wake-promoting and procognitive activity of modafinil in rodents and healthy volunteers. The aim of this study is to investigate changes in glucose cerebral metabolism in rodents, induced by the combination of modafinil+flecainide low dose (called THN102). Methods The impact of THN102 on brain glucose metabolism was noninvasively investigated using 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography imaging in Sprague-Dawley male rats. Animals were injected with vehicle, flecainide, modafinil, or THN102 and further injected with 18F-2-fluoro-2-deoxy-D-glucose followed by 60-minute Positron Emission Tomography acquisition. 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography images were coregistered to a rat brain template and normalized from the total brain Positron Emission Tomography signal. Voxel-to-voxel analysis was performed using SPM8 software. Comparison of brain glucose metabolism between groups was then performed. Results THN102 significantly increased regional brain glucose metabolism as it resulted in large clusters of 18F-2-fluoro-2-deoxy-D-glucose uptake localized in the cortex, striatum, and amygdala compared with control or drugs administered alone. These regions, highly involved in the regulation of sleep-wake cycle, emotions, and cognitive functions were hence quantitatively modulated by THN102. Conclusion Data presented here provide the first evidence of a regional brain activation induced by THN102, currently being tested in a phase II clinical trial in narcoleptic patients.
Collapse
Affiliation(s)
- Dominique Vodovar
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | | | - Catriona Wimberley
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Claire Leroy
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Géraldine Pottier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Yves Dauvilliers
- National Reference Centre for Narcolepsy, CHU Montpellier, INSERM, France
| | - Christian Giaume
- Collège de France, Centre for Interdisciplinary Research in Biology, Paris, France
| | - Jian-Sheng Lin
- Laboratory WAKING, CRNL-INSERM U1028-CNRS UMR 5292-UCBL, Lyon, France
| | | | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | | |
Collapse
|
12
|
González B, Jayanthi S, Gomez N, Torres OV, Sosa MH, Bernardi A, Urbano FJ, García-Rill E, Cadet JL, Bisagno V. Repeated methamphetamine and modafinil induce differential cognitive effects and specific histone acetylation and DNA methylation profiles in the mouse medial prefrontal cortex. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:1-11. [PMID: 29247759 PMCID: PMC6983674 DOI: 10.1016/j.pnpbp.2017.12.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/10/2017] [Indexed: 11/28/2022]
Abstract
Methamphetamine (METH) and modafinil are psychostimulants with different long-term cognitive profiles: METH is addictive and leads to cognitive decline, whereas modafinil has little abuse liability and is a cognitive enhancer. Increasing evidence implicates epigenetic mechanisms of gene regulation behind the lasting changes that drugs of abuse and other psychotropic compounds induce in the brain, like the control of gene expression by histones 3 and 4 tails acetylation (H3ac and H4ac) and DNA cytosine methylation (5-mC). Mice were treated with a seven-day repeated METH, modafinil or vehicle protocol and evaluated in the novel object recognition (NOR) test or sacrificed 4days after last injection for molecular assays. We evaluated total H3ac, H4ac and 5-mC levels in the medial prefrontal cortex (mPFC), H3ac and H4ac promotor enrichment (ChIP) and mRNA expression (RT-PCR) of neurotransmitter systems involved in arousal, wakefulness and cognitive control, like dopaminergic (Drd1 and Drd2), α-adrenergic (Adra1a and Adra1b), orexinergic (Hcrtr1 and Hcrtr2), histaminergic (Hrh1 and Hrh3) and glutamatergic (AMPA Gria1 and NMDA Grin1) receptors. Repeated METH and modafinil treatment elicited different cognitive outcomes in the NOR test, where modafinil-treated mice performed as controls and METH-treated mice showed impaired recognition memory. METH-treated mice also showed i) decreased levels of total H3ac and H4ac, and increased levels of 5-mC, ii) decreased H3ac enrichment at promoters of Drd2, Hcrtr1/2, Hrh1 and Grin1, and increased H4ac enrichment at Drd1, Hrh1 and Grin1, iii) increased mRNA of Drd1a, Grin1 and Gria1. Modafinil-treated mice shared none of these effects and showed increased H3ac enrichment and mRNA expression at Adra1b. Modafinil and METH showed similar effects linked to decreased H3ac in Hrh3, increased H4ac in Hcrtr1, and decreased mRNA expression of Hcrtr2. The specific METH-induced epigenetic and transcriptional changes described here may be related to the long-term cognitive decline effects of the drug and its detrimental effects on mPFC function. The lack of similar epigenetic effects of chronic modafinil administration supports this notion.
Collapse
Affiliation(s)
- Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, Maryland, United States of America
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Oscar V. Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, California, United States of America
| | - Máximo H. Sosa
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco J. Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Edgar García-Rill
- Center for Translational Neuroscience, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, Maryland, United States of America.,Corresponding authors: Veronica Bisagno, Ph.D. Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Junín 956, piso 5, C1113-Buenos Aires, Argentina. Phone: (+54-11) 4961-6784, Fax: (+54-11) 4963-8593. Jean-Lud Cadet, MD
| | - Verónica Bisagno
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
13
|
Mar AC, Nilsson SRO, Gamallo-Lana B, Lei M, Dourado T, Alsiö J, Saksida LM, Bussey TJ, Robbins TW. MAM-E17 rat model impairments on a novel continuous performance task: effects of potential cognitive enhancing drugs. Psychopharmacology (Berl) 2017; 234:2837-2857. [PMID: 28744563 PMCID: PMC5591806 DOI: 10.1007/s00213-017-4679-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/18/2017] [Indexed: 12/02/2022]
Abstract
RATIONALE Impairments in attention and inhibitory control are endophenotypic markers of neuropsychiatric disorders such as schizophrenia and represent key targets for therapeutic management. Robust preclinical models and assays sensitive to clinically relevant treatments are crucial for improving cognitive enhancement strategies. OBJECTIVES We assessed a rodent model with neural and behavioral features relevant to schizophrenia (gestational day 17 methylazoxymethanol acetate treatment (MAM-E17)) on a novel test of attention and executive function, and examined the impact of putative nootropic drugs. METHODS MAM-E17 and sham control rats were trained on a novel touchscreen-based rodent continuous performance test (rCPT) designed to closely mimic the human CPT paradigm. Performance following acute, systemic treatment with an array of pharmacological compounds was investigated. RESULTS Two cohorts of MAM-E17 rats were impaired on rCPT performance including deficits in sensitivity (d') and increased false alarm rates (FARs). Sulpiride (0-30 mg/kg) dose-dependently reduced elevated FAR in MAM-E17 rats whereas low-dose modafinil (8 mg/kg) only improved d' in sham controls. ABT-594 (5.9-19.4 μg/kg) and modafinil (64 mg/kg) showed expected stimulant-like effects, while LSN2463359 (5 mg/kg), RO493858 (10 mg/kg), atomoxetine (0.3-1 mg/kg), and sulpiride (30 mg/kg) showed expected suppressant effects on performance across all animals. Donepezil (0.1-1 mg/kg) showed near-significant enhancements in d', and EVP-6124 (0.3-3 mg/kg) exerted no effects in the rCPT paradigm. CONCLUSION The MAM-E17 model exhibits robust and replicable impairments in rCPT performance that resemble attention and inhibitory control deficits seen in schizophrenia. Pharmacological profiles were highly consistent with known drug effects on cognition in preclinical and clinical studies. The rCPT is a sensitive and reliable tool with high translational potential for understanding the etiology and treatment of disorders affecting attention and executive dysfunction.
Collapse
Affiliation(s)
- Adam C Mar
- Neuroscience Institute, New York University Medical Center, New York, NY, 10016, USA.
- Department of Neuroscience and Physiology, New York University Medical Center, New York, NY, USA.
- Department of Psychology, University of Cambridge, Cambridge, UK.
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| | - Simon R O Nilsson
- Neuroscience Institute, New York University Medical Center, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Medical Center, New York, NY, USA
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Begoña Gamallo-Lana
- Neuroscience Institute, New York University Medical Center, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Medical Center, New York, NY, USA
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Ming Lei
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Health Industry Management, Beijing International Studies University, 1 Dingfuzhuang Nanli, Beijing, China
| | - Theda Dourado
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Johan Alsiö
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Neuroscience, Unit of Functional Neurobiology, University of Uppsala, Uppsala, Sweden
| | - Lisa M Saksida
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Molecular Medicine Research Group, Robarts Research Institute, Western University, London, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- The Brain and Mind Institute, Western University, London, ON, Canada
| | - Timothy J Bussey
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Molecular Medicine Research Group, Robarts Research Institute, Western University, London, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- The Brain and Mind Institute, Western University, London, ON, Canada
| | - Trevor W Robbins
- Department of Psychology, University of Cambridge, Cambridge, UK
- MRC and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Brain-wide Mapping of Endogenous Serotonergic Transmission via Chemogenetic fMRI. Cell Rep 2017; 21:910-918. [DOI: 10.1016/j.celrep.2017.09.087] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/17/2017] [Accepted: 09/26/2017] [Indexed: 11/18/2022] Open
|
15
|
Galbusera A, De Felice A, Girardi S, Bassetto G, Maschietto M, Nishimori K, Chini B, Papaleo F, Vassanelli S, Gozzi A. Intranasal Oxytocin and Vasopressin Modulate Divergent Brainwide Functional Substrates. Neuropsychopharmacology 2017; 42:1420-1434. [PMID: 27995932 PMCID: PMC5436116 DOI: 10.1038/npp.2016.283] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 11/25/2016] [Accepted: 12/13/2016] [Indexed: 12/20/2022]
Abstract
The neuropeptides oxytocin (OXT) and vasopressin (AVP) have been identified as modulators of emotional social behaviors and associated with neuropsychiatric disorders characterized by social dysfunction. Experimental and therapeutic use of OXT and AVP via the intranasal route is the subject of extensive clinical research. However, the large-scale functional substrates directly engaged by these peptides and their functional dynamics remain elusive. By using cerebral blood volume (CBV) weighted fMRI in the mouse, we show that intranasal administration of OXT rapidly elicits the transient activation of cortical regions and a sustained activation of hippocampal and forebrain areas characterized by high oxytocin receptor density. By contrast, intranasal administration of AVP produced a robust and sustained deactivation in cortico-parietal, thalamic and mesolimbic regions. Importantly, intravenous administration of OXT and AVP did not recapitulate the patterns of modulation produced by intranasal dosing, supporting a central origin of the observed functional changes. In keeping with this notion, hippocampal local field potential recordings revealed multi-band power increases upon intranasal OXT administration. We also show that the selective OXT-derivative TGOT reproduced the pattern of activation elicited by OXT and that the deletion of OXT receptors does not affect AVP-mediated deactivation. Collectively, our data document divergent modulation of brainwide neural systems by intranasal administration of OXT and AVP, an effect that involves key substrates of social and emotional behavior. The observed divergence calls for a deeper investigation of the systems-level mechanisms by which exogenous OXT and AVP modulate brain function and exert their putative therapeutic effects.
Collapse
Affiliation(s)
- Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy
| | - Stefano Girardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giacomo Bassetto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marta Maschietto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Katsuhiko Nishimori
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Bice Chini
- CNR, Institute of Neuroscience, Milan, Italy,Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Francesco Papaleo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN), Italy,Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto (TN) 38068, Italy, Tel: +39 04648028701, E-mail:
| |
Collapse
|
16
|
Luo T, Wang Y, Qin J, Liu ZG, Liu M. Histamine H3 Receptor Antagonist Prevents Memory Deficits and Synaptic Plasticity Disruption Following Isoflurane Exposure. CNS Neurosci Ther 2017; 23:301-309. [PMID: 28168839 DOI: 10.1111/cns.12675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Exposure to pharmacological concentration of inhaled anesthetics such as isoflurane can cause short- or long-term cognitive impairments in preclinical studies. The selective antagonists of the histamine H3 receptors are considered as a promising group of novel therapeutic agents for the treatment of cognitive disorders. In this study, we investigated whether ciproxifan, a nonimidazole antagonist of H3 histamine receptors, could overcome the functional and electrophysiological sequela associated with isoflurane anesthesia. METHODS Adult male Sprague Dawley rats were exposed to 1.4% isoflurane or vehicle gas for 2 h. The memory tests (novel object recognition and passive avoidance) as well as in vivo hippocampal excitatory synaptic potentials were recorded 24 h postanesthesia. Locomotor activity, anxiety, and nociception 24 h after isoflurane were also examined. The drugs (ciproxifan 3 mg/kg or saline) were intraperitoneally injected 30 min prior to the behavioral tests or long-term potentiation induction. RESULTS Animals that were previously (24 h) exposed to 1.4% isoflurane for 2 h displayed no preference for novel objects and had impaired retention of a passive avoidance response at 1 h after sample phase. Treating isoflurane-exposed rats with ciproxifan significantly improved the memory performance, as evidenced by an increased discrimination ratio in objects recognition and prolonged retention time in passive avoidance test. Accordingly, hippocampus long-term potentiation was reduced in animals that received isoflurane, while administration of ciproxifan completely abolished the effect of isoflurane exposure on synaptic plasticity. Neither isoflurane nor ciproxifan altered motor performance, anxiety, and nociceptive responses. CONCLUSION These results suggest that H3R in the CNS, probably in the hippocampus, may serve as therapeutic target for improvement of anesthesia-associated cognitive deficits.
Collapse
Affiliation(s)
- Tao Luo
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ying Wang
- Materials Characterization and Preparation Center, Southern University of Science and Technology, Shenzhen, China
| | - Jian Qin
- Central Laboratory, Wuhan University Renmin Hospital, Wuhan, China
| | - Zhi-Gang Liu
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Min Liu
- Central Laboratory, Wuhan University Renmin Hospital, Wuhan, China
| |
Collapse
|
17
|
Susta M, Nemcova V, Bizik G, Sonka K. Emotion stimulus processing in narcolepsy with cataplexy. J Sleep Res 2016; 26:30-37. [DOI: 10.1111/jsr.12444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Marek Susta
- St Elisabeth University of Health and Social Sciences; Bratislava Slovakia
- 1st Faculty of Medicine; Department of Neurology; Charles University and General University Hospital; Prague 2 Czech Republic
| | - Veronika Nemcova
- 1st Faculty of Medicine; Department of Anatomy; Charles University; Prague 2 Czech Republic
| | - Gustav Bizik
- Department of Psychiatry; Aalborg University Hospital; Aalborg Denmark
| | - Karel Sonka
- 1st Faculty of Medicine; Department of Neurology; Charles University and General University Hospital; Prague 2 Czech Republic
| |
Collapse
|
18
|
Duchêne A, Perier M, Zhao Y, Liu X, Thomasson J, Chauveau F, Piérard C, Lagarde D, Picoli C, Jeanson T, Mouthon F, Dauvilliers Y, Giaume C, Lin JS, Charvériat M. Impact of Astroglial Connexins on Modafinil Pharmacological Properties. Sleep 2016; 39:1283-92. [PMID: 27091533 DOI: 10.5665/sleep.5854] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/27/2016] [Indexed: 01/11/2023] Open
Abstract
STUDY OBJECTIVES Modafinil is a non-amphetaminic wake-promoting compound used as therapy against sleepiness and narcolepsy. Its mode of action is complex, but modafinil has been recently proposed to act as a cellular-coupling enhancer in glial cells, through modulation of gap junctions constituted by connexins. The present study investigated in mice the impact of connexins on the effects of modafinil using connexin inhibitors. METHODS Modafinil was administered alone or combined with inhibitors of astrocyte connexin, meclofenamic acid, or flecainide, respectively, acting on Cx30 and Cx43. Sleep-wake states were monitored in wild-type and narcoleptic orexin knockout mice. A spontaneous alternation task was used to evaluate working memory in wild-type mice. The effects of the compounds on astroglial intercellular coupling were determined using dye transfer in acute cortical slices. RESULTS Meclofenamic acid had little modulation on the effects of modafinil, but flecainide enhanced the wake-promoting and pro-cognitive effects of modafinil. Co-administration of modafinil/flecainide resulted in a marked decrease in the number and duration of direct transitions to rapid eye movement sleep, which are characteristic of narcoleptic episodes in orexin knockout mice. Furthermore, modafinil enhanced the connexin-mediated astroglial cell coupling, whereas flecainide reduced it. Finally, this modafinil-induced effect was reversed by co-administration with flecainide. CONCLUSIONS Our study indicates that flecainide impacts the pharmacological effects of modafinil, likely through the normalization of Cx30-dependent gap junctional coupling in astroglial networks. The enhancement of the wake-promoting, behavioral, and cognitive outcomes of modafinil demonstrated here with flecainide would open new perspectives in the management of sleep disorders such as narcolepsy. COMMENTARY A commentary on this article appears in this issue on page 1175.
Collapse
Affiliation(s)
| | - Magali Perier
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | - Yan Zhao
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | - Xinhe Liu
- Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | - Julien Thomasson
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Frédéric Chauveau
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | | | - Didier Lagarde
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Christèle Picoli
- Theranexus, Lyon, France.,CEA/IMETI/Theranexus, Fontenayaux-Roses, France
| | - Tiffany Jeanson
- Theranexus, Lyon, France.,Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | | | - Yves Dauvilliers
- National Reference Centre for Narcolepsy, Sleep Unit, CHU Montpellier, INSERM U1061, France
| | - Christian Giaume
- Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | - Jian-Sheng Lin
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | | |
Collapse
|
19
|
Cognitive enhancers versus addictive psychostimulants: The good and bad side of dopamine on prefrontal cortical circuits. Pharmacol Res 2016; 109:108-18. [PMID: 26826399 DOI: 10.1016/j.phrs.2016.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 12/19/2022]
Abstract
In this review we describe how highly addictive psychostimulants such as cocaine and methamphetamine actions might underlie hypoexcitabilty in frontal cortical areas observed in clinical and preclinical models of psychostimulant abuse. We discuss new mechanisms that describe how increments on synaptic dopamine release are linked to reduce calcium influx in both pre and postsynaptic compartments on medial PFC networks, therefore modulating synaptic integration and information. Sustained DA neuromodulation by addictive psychostimulants can "lock" frontal cortical networks in deficient states. On the other hand, other psychostimulants such as modafinil and methylphenidate are considered pharmacological neuroenhancement agents that are popular among healthy people seeking neuroenhancement. More clinical and preclinical research is needed to further clarify mechanisms of actions and physiological effects of cognitive enhancers which show an opposite pattern compared to chronic effect of addictive psychostimulants: they appear to increase cortical excitability. In conclusion, studies summarized here suggest that there is frontal cortex hypoactivity and deficient inhibitory control in drug-addicted individuals. Thus, additional research on physiological effects of cognitive enhancers like modafinil and methylphenidate seems necessary in order to expand current knowledge on mechanisms behind their therapeutic role in the treatment of addiction and other neuropsychiatric disorders.
Collapse
|
20
|
Limbic thalamus and state-dependent behavior: The paraventricular nucleus of the thalamic midline as a node in circadian timing and sleep/wake-regulatory networks. Neurosci Biobehav Rev 2015; 54:3-17. [DOI: 10.1016/j.neubiorev.2014.11.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/09/2014] [Accepted: 11/21/2014] [Indexed: 12/21/2022]
|
21
|
Lepelletier FX, Tauber C, Nicolas C, Solinas M, Castelnau P, Belzung C, Emond P, Cortese S, Faraone SV, Chalon S, Galineau L. Prenatal exposure to methylphenidate affects the dopamine system and the reactivity to natural reward in adulthood in rats. Int J Neuropsychopharmacol 2014; 18:pyu044. [PMID: 25522388 PMCID: PMC4360227 DOI: 10.1093/ijnp/pyu044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 06/24/2014] [Accepted: 07/08/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Methylphenidate (MPH) is a commonly-used medication for the treatment of children with Attention-Deficit/Hyperactivity Disorders (ADHD). However, its prescription to adults with ADHD and narcolepsy raises the question of how the brain is impacted by MPH exposure during pregnancy. The goal of this study was to elucidate the long-term neurobiological consequences of prenatal exposure to MPH using a rat model. METHODS We focused on the effects of such treatment on the adult dopamine (DA) system and on the reactivity of animals to natural rewards. RESULTS This study shows that adult male rats prenatally exposed to MPH display elevated expression of presynaptic DA markers in the DA cell bodies and the striatum. Our results also suggest that MPH-treated animals could exhibit increased tonic DA activity in the mesolimbic pathway, altered signal-to-noise ratio after a pharmacological stimulation, and decreased reactivity to the locomotor effects of cocaine. Finally, we demonstrated that MPH rats display a decreased preference and motivation for sucrose. CONCLUSIONS This is the first preclinical study reporting long-lasting neurobiological alterations of DA networks as well as alterations in motivational behaviors for natural rewards after a prenatal exposure to MPH. These results raise concerns about the possible neurobiological consequences of MPH treatment during pregnancy.
Collapse
Affiliation(s)
- François-Xavier Lepelletier
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Clovis Tauber
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Céline Nicolas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Marcello Solinas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Pierre Castelnau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Catherine Belzung
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Patrick Emond
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Samuele Cortese
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Stephen V Faraone
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Sylvie Chalon
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Laurent Galineau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone).
| |
Collapse
|
22
|
Raineri M, González B, Rivero-Echeto C, Muñiz JA, Gutiérrez ML, Ghanem CI, Cadet JL, García-Rill E, Urbano FJ, Bisagno V. Differential effects of environment-induced changes in body temperature on modafinil's actions against methamphetamine-induced striatal toxicity in mice. Neurotox Res 2014; 27:71-83. [PMID: 25261212 DOI: 10.1007/s12640-014-9493-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/18/2014] [Accepted: 09/22/2014] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) exposure can produce hyperthermia that might lead to toxicity and death. Modafinil is a wake-promoting compound that is also been prescribed off-label to treat METH dependence. Modafinil has shown neuroprotective properties against METH harmful effects in animal models. The goal of the present study was to test if the prevention of hyperthermia might play a role on the neuroprotective actions of modafinil against METH toxicity using various ambient temperatures. METH was administered to female C57BL/6 mice in a binge regimen: 4 × 5 mg/kg, 2 h apart; modafinil (90 mg/kg) was injected twice, 1 h before first and fourth METH injections. Drugs were given at cold ambient temperature (14 °C) or hot ambient temperature (29 °C). Body temperature was measured during treatments. Brains were dissected out 6 days after treatments and processed for tyrosine hydroxylase (TH), dopamine transporter (DAT), GFAP and c-Fos immunohistochemistry. Exposure to hot ambient temperature exacerbated METH toxicity evidenced by striatal reductions in TH and DAT and increased GFAP immmunoreactivity. Modafinil counteracted reductions in TH and DAT, but failed to block astroglial activation. At both ambient temperatures tested modafinil did induce increments in GFAP, but the magnitude was significantly lower than the one induced by METH. Both drugs induced increases in c-Fos positive nuclei; modafinil did not block this effect. Our results suggest that protective effects of modafinil against METH-induced neurotoxicity may be dependent, in part, to its hypothermic effects. Nevertheless, modafinil maintained some protective properties on METH-induced alterations in the striatum at different ambient temperatures.
Collapse
Affiliation(s)
- Mariana Raineri
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (ININFA-UBA-CONICET), Ciudad Autónoma de Buenos Aires, Junín 956, piso 5, C1113, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Turner C, Belyavin AJ, Nicholson AN. Duration of activity and mode of action of modafinil: Studies on sleep and wakefulness in humans. J Psychopharmacol 2014; 28:643-54. [PMID: 24306135 DOI: 10.1177/0269881113508173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The duration of activity of modafinil was investigated in healthy male volunteers in two double-blind crossover studies. Mode of action was explored using a statistical model concerned with the relationship between total sleep duration and that of rapid eye movement (REM) sleep. Nocturnal sleep (23:00-07:00) followed by next-day performance (09:00-17:00) was studied in 12 subjects administered 100, 200, 300 mg modafinil and placebo, 0.5 h before bedtime. Performance overnight (19:00-08:45) followed by sleep (09:15-15:15) was studied in nine subjects administered 100, 200, 300, 400 mg modafinil, 300 mg caffeine and placebo at 22:15. Modafinil dose-dependently reduced sleep duration (nocturnal: 200 mg, p<0.05; 300 mg, p<0.001; morning: 300 and 400 mg, p<0.05) and REM sleep (nocturnal: 300 mg; morning: 400 mg; p<0.05). The statistical model revealed that reduced REM sleep was due to alerting activity, with no evidence of direct suppression of REM sleep, suggesting dopaminergic activity. Enhanced performance with modafinil during overnight work varied with dose (200 mg>100 mg; 300, 400 mg>200, 100 mg, caffeine). However, in the study of next-day performance, the enhancement was attenuated at the highest dose (300 mg) by the greater disturbance of prior sleep. These findings indicate that modafinil has a long duration of action, with alerting properties arising predominantly from dopaminergic activity.
Collapse
Affiliation(s)
- C Turner
- Lately: QinetiQ Ltd, Farnborough, UK
| | | | - A N Nicholson
- Lately: Centre for Human and Aerospace Physiological Sciences, King's College London, London, UK
| |
Collapse
|
24
|
Minzenberg MJ, Gomes GC, Yoon JH, Watrous AJ, Geng J, Firl AJ, Carter CS. Modafinil augments oscillatory power in middle frequencies during rule selection. Psychophysiology 2014; 51:510-9. [PMID: 24611660 DOI: 10.1111/psyp.12201] [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: 07/14/2013] [Accepted: 01/17/2014] [Indexed: 01/05/2023]
Abstract
Control-related cognitive processes are associated with cortical oscillations and modulated by catecholamine neurotransmitters. It remains unclear how catecholamine systems modulate control-related oscillations. We tested modafinil effects on rule-related 4-30 Hz oscillations, with double-blind, placebo-controlled (within-subjects) testing of 22 healthy adults, using EEG during cognitive control task performance. EEG data underwent time-frequency decomposition with Morlet wavelets to determine power of 4-30 Hz oscillations. Modafinil enhanced oscillatory power associated with high-control rule selection in theta, alpha, and beta ranges, with a frontotemporal topography and minimal effects during rule maintenance. Augmentation of catecholamine signaling enhances middle-frequency cortical oscillatory power associated with rule selection, which may subserve diverse subcomponent processes in proactive cognitive control.
Collapse
Affiliation(s)
- Michael J Minzenberg
- Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, California, USA; San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Modafinil improves methamphetamine-induced object recognition deficits and restores prefrontal cortex ERK signaling in mice. Neuropharmacology 2014; 87:188-97. [PMID: 24530829 DOI: 10.1016/j.neuropharm.2014.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 01/29/2014] [Accepted: 02/04/2014] [Indexed: 11/21/2022]
Abstract
Chronic use of methamphetamine (METH) leads to long-lasting cognitive dysfunction in humans and in animal models. Modafinil is a wake-promoting compound approved for the treatment of sleeping disorders. It is also prescribed off label to treat METH dependence. In the present study, we investigated whether modafinil could improve cognitive deficits induced by sub-chronic METH treatment in mice by measuring visual retention in a Novel Object Recognition (NOR) task. After sub-chronic METH treatment (1 mg/kg, once a day for 7 days), mice performed the NOR task, which consisted of habituation to the object recognition arena (5 min a day, 3 consecutive days), training session (2 equal objects, 10 min, day 4), and a retention session (1 novel object, 5 min, day 5). One hour before the training session, mice were given a single dose of modafinil (30 or 90 mg/kg). METH-treated mice showed impairments in visual memory retention, evidenced by equal preference of familiar and novel objects during the retention session. The lower dose of modafinil (30 mg/kg) had no effect on visual retention scores in METH-treated mice, while the higher dose (90 mg/kg) rescued visual memory retention to control values. We also measured extracellular signal-regulated kinase (ERK) phosphorylation in medial prefrontal cortex (mPFC), hippocampus, and nucleus accumbens (NAc) of METH- and vehicle-treated mice that received modafinil 1 h before exposure to novel objects in the training session, compared to mice placed in the arena without objects. Elevated ERK phosphorylation was found in the mPFC of vehicle-treated mice, but not in METH-treated mice, exposed to objects. The lower dose of modafinil had no effect on ERK phosphorylation in METH-treated mice, while 90 mg/kg modafinil treatment restored the ERK phosphorylation induced by novelty in METH-treated mice to values comparable to controls. We found neither a novelty nor treatment effect on ERK phosphorylation in hippocampus or NAc of vehicle- and METH-treated mice receiving acute 90 mg/kg modafinil treatment. Our results showed a palliative role of modafinil against METH-induced visual cognitive impairments, possibly by normalizing ERK signaling pathways in mPFC. Modafinil may be a valuable pharmacological tool for the treatment of cognitive deficits observed in human METH abusers as well as in other neuropsychiatric conditions. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
26
|
Sforazzini F, Schwarz AJ, Galbusera A, Bifone A, Gozzi A. Distributed BOLD and CBV-weighted resting-state networks in the mouse brain. Neuroimage 2014; 87:403-15. [DOI: 10.1016/j.neuroimage.2013.09.050] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/14/2013] [Accepted: 09/22/2013] [Indexed: 01/14/2023] Open
|
27
|
Chen CR, Yang SR, Liu YY, Qu WM, Urade Y, Huang ZL. Roles of adrenergic α1 and dopamine D1 and D2 receptors in the mediation of the desynchronization effects of modafinil in a mouse EEG synchronization model. PLoS One 2013; 8:e76102. [PMID: 24116090 DOI: 10.1371/journal.pone.0076102] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 08/20/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Synchronized electroencephalogram (EEG) activity is observed in pathological stages of cognitive impairment and epilepsy. Modafinil, known to increase the release of catecholamines, is a potent wake-promoting agent, and has shown some abilities to desynchronize EEG,but its receptor mechanisms by which modafinil induces desynchoronization remain to be elucidated. Here we used a pharmacological EEG synchronization model to investigate the involvement of adrenergic α1 receptors (R, α1R) and dopamine (DA) D1 and D2 receptors (D1Rs and D2Rs) on modafinil-induced desynchronization in mice. METHODOLOGY/PRINCIPAL FINDINGS Mice were treated with cholinergic receptor antagonist scopolamine and monoamine depletor reserpine to produce experimental EEG synchronization characterized by continuous large-amplitude synchronized activity, with prominent increased delta and decreased theta, alpha, and beta power density. The results showed that modafinil produced an EEG desynchronization in the model. This was characterized by a general decrease in amplitude of all the frequency bands between 0 and 20 Hz, a prominent reduction in delta power density, and an increase in theta power density. Adrenergic α1R antagonist terazosin (1 mg/kg, i.p.) completely antagonized the EEG desynchronization effects of modafinil at 90 mg/kg. However, DA D1R and D2R blockers partially attenuated the effects of modafinil. The modafinil-induced decrease in the amplitudes of the delta, theta, alpha, and beta waves and in delta power density were completely abolished by pretreatment with a combination of the D1R antagonist SCH 23390 (30 µg/kg) and the D2R antagonist raclopride (2 mg/kg, i.p.). CONCLUSIONS/SIGNIFICANCE These results suggest that modafinil-mediated desynchronization may be attributed to the activation of adrenergic α1R, and dopaminergic D1R and D2R in a model of EEG synchronization.
Collapse
Affiliation(s)
- Chang-Rui Chen
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
28
|
Differential effect of orexin-1 and CRF-1 antagonism on stress circuits: a fMRI study in the rat with the pharmacological stressor Yohimbine. Neuropsychopharmacology 2013; 38:2120-30. [PMID: 23736277 PMCID: PMC3773661 DOI: 10.1038/npp.2013.109] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/22/2013] [Accepted: 05/03/2013] [Indexed: 12/23/2022]
Abstract
Translational approaches to study the neural substrates of stress and assess the mechanistic efficacy of novel anti-anxiety agents necessitate the use of stressors with a similar degree of saliency across species. The alpha-2 adrenoreceptor antagonist yohimbine represents an attractive experimental tool owing to its well-documented stress-inducing properties in humans and laboratory species. We investigated the neural substrates engaged by yohimbine in the rat brain by using functional magnetic resonance imaging and mapped their modulation by neurotransmitter systems involved in stress responses. Yohimbine elicited a composite pattern of brain activation, highlighting the recruitment of cortico-striato-thalamic regions and extra-hypothalamic stress neurocircuits. This effect was strongly attenuated by the α-2-adrenoceptor agonist medetomidine and by the dopamine (DA) D1 receptor antagonist SCH23390, thus revealing a primary contribution of both norepinephrine and DA on the neurofunctional cascade elicited by the drug. Pretreatment with the corticotrophin-releasing factor type-1 receptor (CRF1R) antagonist CP154,526 produced a region-dependent inhibition of yohimbine-induced activation in the amygdala, striatum, and cingulate cortex, while the orexin type-1 receptor (OX1R) antagonists GSK1059865 robustly inhibited the response in fronto-hippocampal regions as well as in several key components of the extended amygdala. CP154,526 and GSK1059865 did not prevent yohimbine-induced plasma corticosterone release, a finding that corroborates a central origin of the effects mapped. Our findings provide novel insight into the brain substrates and neurochemical mediators engaged by the stress-inducing agent yohimbine. The differential pattern of inhibition produced by CRF1R and OX1R antagonists suggests that these two neuropeptide systems can modulate the functional response to stress via distinct central neural pathways.
Collapse
|
29
|
Mereu M, Bonci A, Newman AH, Tanda G. The neurobiology of modafinil as an enhancer of cognitive performance and a potential treatment for substance use disorders. Psychopharmacology (Berl) 2013; 229:415-34. [PMID: 23934211 PMCID: PMC3800148 DOI: 10.1007/s00213-013-3232-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/28/2013] [Indexed: 12/31/2022]
Abstract
RATIONALE AND OBJECTIVES Modafinil (MOD) and its R-enantiomer (R-MOD) are approved medications for narcolepsy and other sleep disorders. They have also been used, off-label, as cognitive enhancers in populations of patients with mental disorders, including substance abusers that demonstrate impaired cognitive function. A debated nonmedical use of MOD in healthy individuals to improve intellectual performance is raising questions about its potential abuse liability in this population. RESULTS AND CONCLUSIONS MOD has low micromolar affinity for the dopamine transporter (DAT). Inhibition of dopamine (DA) reuptake via the DAT explains the enhancement of DA levels in several brain areas, an effect shared with psychostimulants like cocaine, methylphenidate, and the amphetamines. However, its neurochemical effects and anatomical pattern of brain area activation differ from typical psychostimulants and are consistent with its beneficial effects on cognitive performance processes such as attention, learning, and memory. At variance with typical psychostimulants, MOD shows very low, if any, abuse liability, in spite of its use as a cognitive enhancer by otherwise healthy individuals. Finally, recent clinical studies have focused on the potential use of MOD as a medication for treatment of drug abuse, but have not shown consistent outcomes. However, positive trends in several result measures suggest that medications that improve cognitive function, like MOD or R-MOD, may be beneficial for the treatment of substance use disorders in certain patient populations.
Collapse
Affiliation(s)
- Maddalena Mereu
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Antonello Bonci
- Synaptic Plasticity Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Amy Hauck Newman
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Gianluigi Tanda
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| |
Collapse
|
30
|
Hooker B, Tobon G, Baker S, Zhu C, Hesterman J, Schmidt K, Rajagovindan R, Chandran P, Joshi S, Bannon A, Hoppin J, Beaver J, Fox G, Day M, Upadhyay J. Gabapentin-induced pharmacodynamic effects in the spinal nerve ligation model of neuropathic pain. Eur J Pain 2013; 18:223-37. [DOI: 10.1002/j.1532-2149.2013.00364.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2013] [Indexed: 12/15/2022]
Affiliation(s)
- B.A. Hooker
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | | | - S.J. Baker
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - C. Zhu
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | | | | | - R. Rajagovindan
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - P. Chandran
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - S.K. Joshi
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | - A.W. Bannon
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | | | - J. Beaver
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - G.B. Fox
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - M. Day
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - J. Upadhyay
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| |
Collapse
|
31
|
Pharmacological imaging as a tool to visualise dopaminergic neurotoxicity. Neuropharmacology 2013; 84:159-69. [PMID: 23851258 DOI: 10.1016/j.neuropharm.2013.06.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 06/06/2013] [Accepted: 06/27/2013] [Indexed: 11/20/2022]
Abstract
Dopamine abnormalities underlie a wide variety of psychopathologies, including ADHD and schizophrenia. A new imaging technique, pharmacological magnetic resonance imaging (phMRI), is a promising non-invasive technique to visualize the dopaminergic system in the brain. In this review we explore the clinical potential of phMRI in detecting dopamine dysfunction or neurotoxicity, assess its strengths and weaknesses and identify directions for future research. Preclinically, phMRI is able to detect severe dopaminergic abnormalities quite similar to conventional techniques such as PET and SPECT. phMRI benefits from its high spatial resolution and the possibility to visualize both local and downstream effects of dopaminergic neurotransmission. In addition, it allows for repeated measurements and assessments in vulnerable populations. The major challenge is the complex interpretation of phMRI results. Future studies in patients with dopaminergic abnormalities need to confirm the currently reviewed preclinical findings to validate the technique in a clinical setting. Eventually, based on the current review we expect that phMRI can be of use in a clinical setting involving vulnerable populations (such as children and adolescents) for diagnosis and monitoring treatment efficacy. This article is part of the Special Issue Section entitled 'Neuroimaging in Neuropharmacology'.
Collapse
|
32
|
Dodd GT, Worth AA, Hodkinson DJ, Srivastava RK, Lutz B, Williams SR, Luckman SM. Central functional response to the novel peptide cannabinoid, hemopressin. Neuropharmacology 2013; 71:27-36. [PMID: 23542442 DOI: 10.1016/j.neuropharm.2013.03.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 02/19/2013] [Accepted: 03/07/2013] [Indexed: 02/05/2023]
Abstract
Hemopressin is the first peptide ligand to be described for the CB₁ cannabinoid receptor. Hemopressin acts as an inverse agonist in vivo and can cross the blood-brain barrier to both inhibit appetite and induce antinociception. Despite being highly effective, synthetic CB₁ inverse agonists are limited therapeutically due to unwanted, over dampening of central reward pathways. However, hemopressin appears to have its effect on appetite by affecting satiety rather than reward, suggesting an alternative mode of action which might avoid adverse side effects. Here, to resolve the neuronal circuitry mediating hemopressin's actions, we have combined blood-oxygen-level-dependent, pharmacological-challenge magnetic resonance imaging with c-Fos functional activity mapping to compare brain regions responsive to systemic administration of hemopressin and the synthetic CB₁ inverse agonist, AM251. Using these complementary methods, we demonstrate that hemopressin activates distinct neuronal substrates within the brain, focused mainly on the feeding-related circuits of the mediobasal hypothalamus and in nociceptive regions of the periaqueductal grey (PAG) and dorsal raphe (DR). In contrast to AM251, there is a distinct lack of activation of the brain reward centres, such as the ventral tegmental area, nucleus accumbens and orbitofrontal cortex, which normally form a functional activity signature for the central action of synthetic CB₁ receptor inverse agonists. Thus, hemopressin modulates the function of key feeding-related brain nuclei of the mediobasal hypothalamus, and descending pain pathways of the PAG and DR, and not higher limbic structures. Thus, hemopressin may offer behaviourally selective effects on nociception and appetite, without engaging reward pathways.
Collapse
Affiliation(s)
- Garron T Dodd
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, UK.
| | | | | | | | | | | | | |
Collapse
|
33
|
Gozzi A, Agosta F, Massi M, Ciccocioppo R, Bifone A. Reduced limbic metabolism and fronto-cortical volume in rats vulnerable to alcohol addiction. Neuroimage 2012; 69:112-9. [PMID: 23261637 DOI: 10.1016/j.neuroimage.2012.12.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 11/25/2022] Open
Abstract
Alcohol abuse is associated with long-term reductions in fronto-cortical volume and limbic metabolism. However, an unanswered question in alcohol research is whether these alterations are the sole consequence of chronic alcohol use, or contain heritable contributions reflecting biological propensity toward ethanol addiction. Animal models of genetic predisposition to alcohol dependence can be used to investigate the role of inborn brain abnormalities in the aetiology of alcoholism. Here we used magnetic resonance imaging (MRI) in the Marchigian-Sardinian (msP) alcohol-preferring rats to assess the presence of inherited structural or functional brain alterations. Alcohol-naïve msP (N=22) and control rats (N=26) were subjected to basal cerebral blood volume (bCBV) mapping followed by voxel-based morphometry (VBM) of grey matter and tract-based spatial statistics mapping of white matter fractional anisotropy. msP rats exhibited significantly reduced bCBV, an established marker of resting brain function, in focal cortico-limbic and thalamic areas, together with reduced grey matter volume in the thalamus, ventral tegmental area, insular and cingulate cortex. No statistically significant differences in fractional anisotropy were observed between groups. These findings highlight the presence of inborn grey matter and metabolic abnormalities in alcohol-naïve msP rats, the localization and sign of which are remarkably similar to those mapped in abstinent alcoholics and subjects at high risk for alcohol dependence. Collectively, these results point for a significant role of heritable neurofunctional brain alterations in biological propensity toward ethanol addiction, and support the translational use of advanced imaging methods to describe the circuital determinants of vulnerability to drug addiction.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Istituto Italiano di Tecnologia, Center for Nanotechnology Innovation, Pisa, & Center for Neuroscience and Cognitive Systems at UniTn, Rovereto, Italy.
| | | | | | | | | |
Collapse
|
34
|
Addiction-related gene regulation: risks of exposure to cognitive enhancers vs. other psychostimulants. Prog Neurobiol 2012; 100:60-80. [PMID: 23085425 DOI: 10.1016/j.pneurobio.2012.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/02/2012] [Accepted: 10/08/2012] [Indexed: 01/09/2023]
Abstract
The psychostimulants methylphenidate (Ritalin, Concerta), amphetamine (Adderall), and modafinil (Provigil) are widely used in the treatment of medical conditions such as attention-deficit hyperactivity disorder and narcolepsy and, increasingly, as "cognitive enhancers" by healthy people. The long-term neuronal effects of these drugs, however, are poorly understood. A substantial amount of research over the past two decades has investigated the effects of psychostimulants such as cocaine and amphetamines on gene regulation in the brain because these molecular changes are considered critical for psychostimulant addiction. This work has determined in some detail the neurochemical and cellular mechanisms that mediate psychostimulant-induced gene regulation and has also identified the neuronal systems altered by these drugs. Among the most affected brain systems are corticostriatal circuits, which are part of cortico-basal ganglia-cortical loops that mediate motivated behavior. The neurotransmitters critical for such gene regulation are dopamine in interaction with glutamate, while other neurotransmitters (e.g., serotonin) play modulatory roles. This review presents (1) an overview of the main findings on cocaine- and amphetamine-induced gene regulation in corticostriatal circuits in an effort to provide a cellular framework for (2) an assessment of the molecular changes produced by methylphenidate, medical amphetamine (Adderall), and modafinil. The findings lead to the conclusion that protracted exposure to these cognitive enhancers can induce gene regulation effects in corticostriatal circuits that are qualitatively similar to those of cocaine and other amphetamines. These neuronal changes may contribute to the addiction liability of the psychostimulant cognitive enhancers.
Collapse
|
35
|
Scoriels L, Jones PB, Sahakian BJ. Modafinil effects on cognition and emotion in schizophrenia and its neurochemical modulation in the brain. Neuropharmacology 2012; 64:168-84. [PMID: 22820555 DOI: 10.1016/j.neuropharm.2012.07.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/03/2012] [Accepted: 07/05/2012] [Indexed: 02/04/2023]
Abstract
Modafinil is a central nervous system wake promoting agent used for the treatment of excessive daytime sleeping. Its vigilance promoting properties and low abuse potential has intrigued the scientific community and has led to use it as a cognitive enhancer, before its neural functions were understood. Here, we review the effects of modafinil in human cognition and emotion and its specific actions on symptoms in patients with schizophrenia and whether these are consistently effective throughout the literature. We also performed a systematic review on the effects of modafinil on neurotransmitter signalling in different areas of the brain in order to better understand the neuromechanisms of its cognitive and emotional enhancing properties. A review of its effects in schizophrenia suggests that modafinil facilitates cognitive functions, with pro-mnemonic effects and problem solving improvements. Emotional processing also appears to be enhanced by the drug, although to date there are only a limited number of studies. The systematic review on the neurochemical modulation of the modafinil suggests that its mnemonic enhancing properties might be the result of glutamatergic and dopaminergic increased neuronal activation in the hippocampus and in the prefrontal cortex respectively. Other neurotransmitters were also activated by modafinil in various limbic brain areas, suggesting that the drug acts on these brain regions to influence emotional responses. These reviews seek to delineate the neuronal mechanisms by which modafinil affects cognitive and emotional function. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
|