1
|
De Risi M, Cusimano L, Bujanda Cundin X, Pizzo M, Gigante Y, Monaco M, Di Eugenio C, De Leonibus E. D1 dopamine receptor antagonists as a new therapeutic strategy to treat autistic-like behaviours in lysosomal storage disorders. Mol Psychiatry 2025:10.1038/s41380-025-02904-6. [PMID: 39865184 DOI: 10.1038/s41380-025-02904-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 12/11/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Lysosomal storage disorders characterized by defective heparan sulfate (HS) degradation, such as Mucopolysaccharidosis type IIIA-D (MPS-IIIA-D), result in neurodegeneration and dementia in children. However, dementia is preceded by severe autistic-like behaviours (ALBs), presenting as hyperactivity, stereotypies, social interaction deficits, and sleep disturbances. The absence of experimental studies on ALBs' mechanisms in MPS-III has led clinicians to adopt symptomatic treatments, such as antipsychotics, which are used for non-genetic neuropsychiatric disorders. However, they have limited efficacy in MPS-III and lead to higher extrapyramidal effects, leaving ALBs in MPS-IIIA as an unmet medical need with a significant burden on patients and their families. Using mouse and cellular models of MPS-IIIA, we have previously shown that ALBs result from increased proliferation of mesencephalic dopamine neurons during embryogenesis. In adulthood, MPS-IIIA mice exhibit an imbalance of dopaminergic receptor subtypes, resulting in striatal overstimulation of the D1 dopamine receptor (D1R)-direct pathway, contrasting with a downregulation of the D2 dopamine receptor (D2R)-indirect pathway. In this study, we aimed to provide an evidence-based pharmacological approach for managing ALBs in MPS-IIIA. We hypothesized that rebalancing dopaminergic receptor signalling with a D1R antagonist, rather than a D2 antagonist, would lead to safe and effective treatment. Neither risperidone nor methylphenidate improves ALBs in the MPS-IIIA mouse model, with the former showing increased cataleptic (extrapyramidal-like) side effects compared to littermate wild-type animals. Methylphenidate, however, showed some beneficial effects on neuroinflammation and later manifesting dementia-like behaviours. In contrast, ecopipam, a D1 antagonist already used in the clinic for other neuropsychiatric disorders, rescues ALBs, cognition, D1 hyperactivity, and does not worsen neurodegenerative signs. These results align with recent evidence highlighting the clinical relevance of D1 antagonists for neuropsychiatric disorders and pave the way for their use in managing psychotic symptoms in neurodegenerative disorders such as dementia with Lewy bodies.
Collapse
Affiliation(s)
- Maria De Risi
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy.
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy.
| | - Lorenzo Cusimano
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Xabier Bujanda Cundin
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mariateresa Pizzo
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Ylenia Gigante
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mariagrazia Monaco
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Chiara Di Eugenio
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy.
- Institute of Biochemistry and Cell Biology, CNR, Via Ramarini 32, Monterotondo Scalo, 00015, Rome, Italy.
| |
Collapse
|
2
|
De Filippo R, Schmitz D. Synthetic surprise as the foundation of the psychedelic experience. Neurosci Biobehav Rev 2024; 157:105538. [PMID: 38220035 PMCID: PMC10839673 DOI: 10.1016/j.neubiorev.2024.105538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Psychedelic agents, such as LSD and psilocybin, induce marked alterations in consciousness via activation of the 5-HT2A receptor (5-HT2ARs). We hypothesize that psychedelics enforce a state of synthetic surprise through the biased activation of the 5-HTRs system. This idea is informed by recent insights into the role of 5-HT in signaling surprise. The effects on consciousness, explained by the cognitive penetrability of perception, can be described within the predictive coding framework where surprise corresponds to prediction error, the mismatch between predictions and actual sensory input. Crucially, the precision afforded to the prediction error determines its effect on priors, enabling a dynamic interaction between top-down expectations and incoming sensory data. By integrating recent findings on predictive coding circuitry and 5-HT2ARs transcriptomic data, we propose a biological implementation with emphasis on the role of inhibitory interneurons. Implications arise for the clinical use of psychedelics, which may rely primarily on their inherent capacity to induce surprise in order to disrupt maladaptive patterns.
Collapse
Affiliation(s)
- Roberto De Filippo
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany.
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Einstein Center for Neuroscience, 10117 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, 10117 Berlin, Germany; Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
3
|
Ben-Azu B, Adebayo OG, Moke EG, Omogbiya AI, Oritsemuelebi B, Chidebe EO, Umukoro E, Nwangwa EK, Etijoro E, Umukoro E, Mamudu EJ, Chukwuma C. Geraniol attenuates behavioral and neurochemical impairments by inhibitions of HPA-axis and oxido-inflammatory perturbations in mice exposed to post-traumatic stress disorder. J Psychiatr Res 2023; 168:165-175. [PMID: 37913743 DOI: 10.1016/j.jpsychires.2023.10.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/23/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
Geraniol is an acyclic isoprenoid monoterpenoid analogue that has been shown to elicit neuroprotective functions, primarily through its ability to stimulate antioxidant and anti-inflammatory systems. An increase in inflammatory cytokines and oxidative stress exacerbate activation hypothalamic-pituitary-adrenal axis (HPA), leading to neurochemical dysfunction, which has important roles in the pathogenesis of post-traumatic disorder (PTSD), a mental health disorder characterized of post-trauma-induced intense fear. The aim of this study was to evaluate the anti-PTSD-like effects and underlying mechanisms of geraniol against single-prolonged-stress (SPS)-induced PTSD in mice. Following concomitant exposure to SPS (triple-paradigm traumatic events) and isolation for 7 days, mice (n = 9) were treated with geraniol (50 and 100 mg/kg, p.o.) or fluoxetine (10 mg/kg, p.o.) from days 8-21. Mice were assessed for behavioral changes. Neurochemical changes, inflammatory, oxido-nitrergic markers, adrenal weight, serum glucose and corticosterone concentrations were assayed. Geraniol inhibits SPS-induced anxiety- and depressive-like features as well as behavioral despair in the depression paradigms. SPS-induced locomotor and memory impairments were also abated by geraniol treatment similarly to fluoxetine. SPS-induced adrenal hypertrophy and increased blood glucose and corticosterone concentrations, were attenuated by the geraniol treatment. Elevated levels of TNF-α and IL-6, and malondialdehyde, nitrite, acetylcholinesterase enzyme were reduced by geraniol. Geraniol also increased glutathione, superoxide-dismutase, and catalase levels as well as dopamine, serotonin concentrations and GABAergic glutamic acid decarboxylase enzyme activity in the striatum, prefrontal cortex and hippocampus in the PTSD-mice relative to SPS control. In conclusion, geraniol attenuates behavioral impairments and neurochemical dysregulations by inhibitions of HPA-axis and oxido-inflammatory perturbations in mice exposed to PTSD.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria.
| | - Olusegun G Adebayo
- Neurophysiology Unit, Department of Physiology, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port-Harcourt, River State, Nigeria
| | - Emuesiri G Moke
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Adrian I Omogbiya
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Benjamin Oritsemuelebi
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel O Chidebe
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emuesiri Umukoro
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Medicine Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Eze K Nwangwa
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel Etijoro
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Emmanuel Umukoro
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Elizabeth J Mamudu
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Chineye Chukwuma
- DELSU Joint Canada-Israel Neuroscience and Biopsychiatry Laboratory, Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| |
Collapse
|
4
|
Garcia-Marin LM, Mulcahy A, Byrne EM, Medland SE, Wray NR, Chafota F, Lind PA, Martin NG, Hickie IB, Rentería ME, Campos AI. Discontinuation of antidepressant treatment: a retrospective cohort study on more than 20,000 participants. Ann Gen Psychiatry 2023; 22:49. [PMID: 38001492 PMCID: PMC10668351 DOI: 10.1186/s12991-023-00480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Factors influencing antidepressant treatment discontinuation are poorly understood. In the present study, we aimed to estimate the prevalence of antidepressant treatment discontinuation and identify demographic characteristics, psychiatric comorbidities, and specific side effects associated with treatment discontinuation. METHODS We leveraged data from the Australian Genetics of Depression Study (AGDS; N = 20,941) to perform a retrospective cohort study on antidepressant treatment discontinuation. Participants were eligible if they were over 18 years of age, had taken antidepressants in the past 4 years, and provided informed consent. RESULTS Among the ten antidepressants studied, the highest discontinuation rates were observed for Mirtazapine (57.3%) and Amitriptyline (51.6%). Discontinuation rates were comparable across sexes except for Mirtazapine, for which women were more likely to discontinue. The two most common side effects, reduced sexual function and weight gain, were not associated with increased odds of treatment discontinuation. Anxiety, agitation, suicidal thoughts, vomiting, and rashes were associated with higher odds for treatment discontinuation, as were lifetime diagnoses of PTSD, ADHD, and a higher neuroticism score. Educational attainment showed a negative (protective) association with discontinuation across medications. CONCLUSIONS Our study suggests that not all side effects contribute equally to discontinuation. Common side effects such as reduced sexual function and weight gain may not necessarily increase the risk of treatment discontinuation. Side effects linked to discontinuation can be divided into two groups, psychopathology related and allergy/intolerance.
Collapse
Affiliation(s)
- Luis M Garcia-Marin
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| | - Aoibhe Mulcahy
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Enda M Byrne
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Sarah E Medland
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Freddy Chafota
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Penelope A Lind
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas G Martin
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Camperdown, NSW, Australia
| | - Miguel E Rentería
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Adrian I Campos
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Liu MN, Tian XY, Fang T, Wu N, Li H, Li J. Insights into the Involvement and Therapeutic Target Potential of the Dopamine System in the Posttraumatic Stress Disorder. Mol Neurobiol 2023; 60:3708-3723. [PMID: 36933147 DOI: 10.1007/s12035-023-03312-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a neuropsychiatric disease closely related to life-threatening events and psychological stress. Re-experiencing, hyperarousal, avoidance, and numbness are the hallmark symptoms of PTSD, but their underlying neurological processes have not been clearly elucidated. Therefore, the identification and development of drugs for PTSD that targets brain neuronal activities have stalled. Considering that the persistent fear memory induced by traumatic stimulation causes high alertness, high arousal, and cognitive impairment of PTSD symptoms. While the midbrain dopamine system can affect physiological processes such as aversive fear memory learning, consolidation, persistence, and extinction, by altering the functions of the dopaminergic neurons, our viewpoint is that the dopamine system plays a considerable role in the PTSD occurrence and acts as a potential therapeutic target of the disorder. This paper reviews recent findings on the structural and functional connections between ventral tegmental area neurons and the core synaptic circuits involved in PTSD, gene polymorphisms related to the dopamine system that confer susceptibility to clinical PTSD. Moreover, the progress of research on medications that target the dopamine system as PTSD therapies is also discussed. Our goal is to offer some hints for early detection and assist in identifying novel, efficient approaches for treating PTSD.
Collapse
Affiliation(s)
- Meng-Nan Liu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Xiao-Yu Tian
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China.,Medical School of Chinese PLA, Beijing, 100853, China
| | - Ting Fang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Ning Wu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China.
| | - Jin Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China.
| |
Collapse
|
6
|
Punski-Hoogervorst JL, Engel-Yeger B, Avital A. Attention deficits as a key player in the symptomatology of posttraumatic stress disorder: A review. J Neurosci Res 2023; 101:1068-1085. [PMID: 36807926 DOI: 10.1002/jnr.25177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/05/2023] [Accepted: 01/28/2023] [Indexed: 02/22/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating psychiatric disorder characterized by symptoms such as re-experiencing of the psychotrauma and hyperarousal. Although current literature mainly discusses the emotionally related aspects of these symptoms, studies also highlight the relation between re-experiencing, hyperarousability, and attention deficits, which are associated with poorer daily function and reduced quality of life. This review provides a comprehensive analysis of the existing research on attention deficits among adults with PTSD. A systematic search through five databases resulted in the inclusion of 48 peer-reviewed, English-language articles, describing 49 distinct studies. Using a total of 47 different attentional assessment tools, the majority of studies investigated sustained (n = 40), divided (n = 16), or selective (n = 14) attention. A total of 30 studies (61.2%) found significant correlations between PTSD symptoms and attention deficits, and 10 studies (20.4%) found that higher levels of attention deficits were predictive of worse PTSD symptoms. Moreover, neuroimaging results of six (f)MRI and three EEG studies identified various potential neurobiological pathways involved, including (pre)frontal attention networks. Together, the body of research shows that attention deficits in individuals with PTSD are common and occur in surroundings with emotionally neutral stimuli. Nonetheless, current treatment strategies do not target these attentional difficulties. We propose a novel perspective to PTSD diagnosis and treatment strategies based on attention deficits and their relation with top-down regulation of re-experiencing and subsequent other PTSD symptoms.
Collapse
Affiliation(s)
- Janne L Punski-Hoogervorst
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Batya Engel-Yeger
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
7
|
Patel S, Keating BA, Dale RC. Anti-inflammatory properties of commonly used psychiatric drugs. Front Neurosci 2023; 16:1039379. [PMID: 36704001 PMCID: PMC9871790 DOI: 10.3389/fnins.2022.1039379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023] Open
Abstract
Mental health and neurodevelopmental disorders are extremely common across the lifespan and are characterized by a complicated range of symptoms that affect wellbeing. There are relatively few drugs available that target disease mechanisms for any of these disorders. Instead, therapeutics are focused on symptoms and syndromes, largely driven by neurotransmitter hypotheses, such as serotonin or dopamine hypotheses of depression. Emerging evidence suggests that maternal inflammation during pregnancy plays a key role in neurodevelopmental disorders, and inflammation can influence mental health expression across the lifespan. It is now recognized that commonly used psychiatric drugs (anti-depressants, anti-psychotics, and mood stabilizers) have anti-inflammatory properties. In this review, we bring together the human evidence regarding the anti-inflammatory mechanisms for these main classes of psychiatric drugs across a broad range of mental health disorders. All three classes of drugs showed evidence of decreasing levels of pro-inflammatory cytokines, particularly IL-6 and TNF-α, while increasing the levels of the anti-inflammatory cytokine, IL-10. Some studies also showed evidence of reduced inflammatory signaling via nuclear factor- (NF-)κB and signal transducer and activator of transcription (STAT) pathways. As researchers, clinicians, and patients become increasingly aware of the role of inflammation in brain health, it is reassuring that these psychiatric drugs may also abrogate this inflammation, in addition to their effects on neurotransmission. Further studies are required to determine whether inflammation is a driver of disease pathogenesis, and therefore should be a therapeutic target in future clinical trials.
Collapse
Affiliation(s)
- Shrujna Patel
- Faculty of Medicine and Health, Kids Neuroscience Centre, The Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia,Faculty of Medicine and Health, Clinical School, The Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia,Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Brooke A. Keating
- Faculty of Medicine and Health, Kids Neuroscience Centre, The Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia,Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Russell C. Dale
- Faculty of Medicine and Health, Kids Neuroscience Centre, The Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia,Faculty of Medicine and Health, Clinical School, The Children's Hospital at Westmead, University of Sydney, Westmead, NSW, Australia,Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW, Australia,*Correspondence: Russell C. Dale ✉
| |
Collapse
|
8
|
Grossman A, Avital A. Emotional and sensory dysregulation as a possible missing link in attention deficit hyperactivity disorder: A review. Front Behav Neurosci 2023; 17:1118937. [PMID: 36935890 PMCID: PMC10017514 DOI: 10.3389/fnbeh.2023.1118937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a common developmental disorder affecting 5-7% of adults and children. We surveyed the literature to examine ADHD through three pillars: developmental characteristics, symptomatology, and treatment strategies. Firstly, in terms of developmental characterstics, early life stress may increase the risk of developing ADHD symptoms according to animal models' research. Secondly, the current core symptoms of ADHD are comprised of inattention, hyperactivity, and impulsivity. However, the up-to-date literature indicates individuals with ADHD experience emotional and sensory dysregulation as well, which early-life stress may also increase the risk of. Finally, we discuss the therapeutic benefits of methylphenidate on both the current core ADHD symptoms and the sensory and emotional dysregulation found in those with ADHD. In summation, we surveyed the recent literature to analyze (i) the potential role of early-life stress in ADHD development, (ii) the involvement of emotional and sensory dysregulation in ADHD symptomatology and finally, (iii) the therapeutic intervention with methylphenidate, aiming to reduce the potential effect of early life stress in ADHD, and mainly emotional and sensory dysregulation. The apparent but currently less recognized additional symptoms of emotional and sensory dysregulation in ADHD call for further investigation of these possible causes and thus increasing treatments efficacy in individuals with ADHD.
Collapse
|
9
|
Kern DM, Teneralli RE, Flores CM, Wittenberg GM, Gilbert JP, Cepeda MS. Revealing Unknown Benefits of Existing Medications to Aid the Discovery of New Treatments for Post‐Traumatic Stress Disorder. PSYCHIATRIC RESEARCH AND CLINICAL PRACTICE 2022; 4:12-20. [PMID: 36101715 PMCID: PMC9175795 DOI: 10.1176/appi.prcp.20210019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/13/2021] [Accepted: 11/20/2021] [Indexed: 11/30/2022] Open
Abstract
Objective To systematically identify novel pharmacological strategies for preventing or treating post‐traumatic stress disorder (PTSD) by leveraging large‐scale analysis of real‐world observational data. Methods Using a self‐controlled study design, the association between 1399 medications and the incidence of PTSD across four US insurance claims databases covering commercially insured, Medicare eligible, and Medicaid patients was examined. A validated algorithm for identifying PTSD in claims data was used, and medications were identified by their RxNorm ingredient. Medications used to treat PTSD or its symptoms (e.g., antidepressants, antipsychotics) were excluded. Medications associated with ≥30% reduction in risk of PTSD in ≥2 databases were identified. Results A total of 137,182,179 individuals were included in the analysis. Fifteen medications met the threshold criteria for a potential protective effect on PTSD; six were categorized as “primary signals” while the remaining nine were considered “potential signals”. The primary signals include a beta blocker that has been previously studied for PTSD, and five medications used to treat attention‐deficit/hyperactivity disorder. The potential signals include four medications used to treat substance use disorders and five medications used to treat sleep disorders. Discussion The medications identified in this analysis provide targets for further research in studies that are designed to examine specific hypotheses regarding these medications and the incidence of PTSD. This work may aid in discovering novel therapeutic approaches to treat PTSD, wherein new and effective treatments are badly needed. Four large US‐based administrative claims databases were used to analyze the association between all marketed prescription medications and the outcome of incident post‐traumatic stress disorder (PTSD) Of the 1399 medications examined, there were 15 that met the strict filtering criteria for showing consistent, moderate‐to‐strong, protective effects against the outcome Medications fell into four main classes: (1) a beta blocker (propranolol), (2) five medications used to treat attention‐deficit/hyperactivity disorder (ADHD), (3) four medications used to treat substance use disorders and (4) five medications used to treat sleep disorders These findings identify rational starting points for future hypothesis‐driven research to explore these associations in greater detail
Collapse
Affiliation(s)
- David M. Kern
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| | - Rachel E. Teneralli
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| | - Christopher M. Flores
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| | - Gayle M. Wittenberg
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| | - James P. Gilbert
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| | - M. Soledad Cepeda
- Janssen Research & Development, Titusville, NJ (D. M. Kern, R. E. Teneralli, G. M. Wittenberg, J. P. Gilbert, M. S. Cepeda); Janssen Research & Development, San Diego (C. M. Flores)
| |
Collapse
|
10
|
Repeated cocaine exposure prior to fear conditioning induces persistency of PTSD-like symptoms and enhancement of hippocampal and amygdala cell density in male rats. Brain Struct Funct 2021; 226:2219-2241. [PMID: 34195855 DOI: 10.1007/s00429-021-02320-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/11/2021] [Indexed: 12/29/2022]
Abstract
Pre- and post-trauma drug use can interfere with recovery from post-traumatic stress disorder (PTSD). However, the biological underpinnings of this interference are poorly understood. Here we examined the effect of pre-fear conditioning cocaine self-administration on PTSD-like symptoms in male rats, and defined impairment of fear extinction as difficulty to recover from PTSD. We also examined cell density changes in brain regions suspected of being involved in resistance to PTSD recovery. Before footshock stress testing, rats were trained to self-administer cocaine during 20 consecutive days, after which they were exposed to footshocks, while other rats continued to self-administer cocaine until the end of the experiment. Upon assessment of three PTSD-like symptoms (fear during situational reminders, anxiety-like behavior, and impairment of recognition memory) and fear extinction learning and memory, changes in cell density were measured in the medial prefrontal cortex, hippocampus, and amygdala. Results show that pre-footshock cocaine exposure did not affect fear during situational reminders. Fear conditioning did not lead to an increase in cocaine consumption. However, in footshock stressed rats, cocaine induced a reduction of anxiety-like behavior, an aggravation of recognition memory decline, and an impairment of extinction memory. These behavioral alterations were associated with increased cell density in the hippocampal CA1, CA2, and CA3 regions and basolateral amygdala, but not in the medial prefrontal cortex. Our findings suggest that enhancement of cell density in the hippocampus and amygdala may be changes associated with drug use, interfering with PTSD recovery.
Collapse
|
11
|
Ullrich D, Mac Gillavry DW. Mini-review: A possible role for galanin in post-traumatic stress disorder. Neurosci Lett 2021; 756:135980. [PMID: 34023414 DOI: 10.1016/j.neulet.2021.135980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/02/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
Several neuroendocrine systems have been implicated in post-traumatic stress disorder, including the mesocortical and mesolimbic dopamine, the norepinephrine, the β-endorphin, the serotonin, and the oxytocin systems. The interaction between these different systems remains, however, largely unknown and a generally accepted unifying theory is thus far lacking. In this review, we suggest that galanergic suppression of dopaminergic neurons in the ventral tegmental may constitute the missing link in a post-traumatic feedback loop. In addition, we address the literature on the negative cross-antagonism in this brain region between the galanin 1 and μ-opioid receptors, which suggests that behavioural patterns which stimulate β-endorphin, a natural μ-opioid receptors ligand, secretion may provide novel avenues for the treatment and prevention of PTSD, as well as for recruitment, training, and leadership processes in high-stress/high-risk professions such as the military, first responders and the police.
Collapse
Affiliation(s)
- David Ullrich
- Department of Military Leadership, University of Defence, Brno, Czech Republic
| | | |
Collapse
|
12
|
Ney LJ, Akhurst J, Bruno R, Laing PAF, Matthews A, Felmingham KL. Dopamine, endocannabinoids and their interaction in fear extinction and negative affect in PTSD. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110118. [PMID: 32991952 DOI: 10.1016/j.pnpbp.2020.110118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
There currently exist few frameworks for common neurobiology between reexperiencing and negative cognitions and mood symptoms of PTSD. Adopting a dopaminergic framework for PTSD unites many aspects of unique symptom clusters, and this approach also links PTSD symptomology to common comorbidities with a common neurobiological deficiency. Here we review the dopamine literature and incorporate it with a growing field of research that describes both the contribution of endocannabinoids to fear extinction and PTSD, as well as the interactions between dopaminergic and endocannabinoid systems underlying this disorder. Based on current evidence, we outline an early, preliminary model that links re-experiencing and negative cognitions and mood in PTSD by invoking the interaction between endocannabinoid and dopaminergic signalling in the brain. These interactions between PTSD, dopamine and endocannabinoids may have implications for future therapies for treatment-resistant and comorbid PTSD patients.
Collapse
Affiliation(s)
- Luke J Ney
- School of Psychology, University of Tasmania, Australia.
| | - Jane Akhurst
- School of Psychology, University of Tasmania, Australia
| | | | - Patrick A F Laing
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Australia
| | | | - Kim L Felmingham
- School of Psychological Sciences, University of Melbourne, Australia
| |
Collapse
|
13
|
Abstract
This case study displays the successful application of Prolonged Exposure (PE) for a client with diagnoses of Posttraumatic Stress Disorder (PTSD), Attention Deficit Hyperactivity Disorder (ADHD), and Generalized Anxiety Disorder (GAD). To our knowledge ADHD has not been examined as a predictor or moderator of PE outcomes. As such, practitioners have precious little information about how to proceed in such cases, which highlights the importance of careful individual assessment and case conceptualization. There is also a dearth of information on the effects of combining PE (a research-supported psychological intervention for PTSD) with psychostimulant medication (a research-supported pharmacological intervention for ADHD). The present case study illustrates a positive synergy between psychostimulant treatment and PE. The unique adjustments made to deliver services (including in the face of COVID-19) are described as well as what this case suggests about the effects of psychostimulant use on PTSD symptoms and the new learning that occurs during PE.
Collapse
|
14
|
Ramon-Duaso C, Gener T, Consegal M, Fernández-Avilés C, Gallego JJ, Castarlenas L, Swanson MS, de la Torre R, Maldonado R, Puig MV, Robledo P. Methylphenidate Attenuates the Cognitive and Mood Alterations Observed in Mbnl2 Knockout Mice and Reduces Microglia Overexpression. Cereb Cortex 2020; 29:2978-2997. [PMID: 30060068 DOI: 10.1093/cercor/bhy164] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 05/25/2018] [Indexed: 12/15/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystem disorder affecting muscle and central nervous system (CNS) function. The cellular mechanisms underlying CNS alterations are poorly understood and no useful treatments exist for the neuropsychological deficits observed in DM1 patients. We investigated the progression of behavioral deficits present in male and female muscleblind-like 2 (Mbnl2) knockout (KO) mice, a rodent model of CNS alterations in DM1, and determined the biochemical and electrophysiological correlates in medial prefrontal cortex (mPFC), striatum and hippocampus (HPC). Male KO exhibited more cognitive impairment and depressive-like behavior than female KO mice. In the mPFC, KO mice showed an overexpression of proinflammatory microglia, increased transcriptional levels of Dat, Drd1, and Drd2, exacerbated dopamine levels, and abnormal neural spiking and oscillatory activities in the mPFC and HPC. Chronic treatment with methylphenidate (MPH) (1 and 3 mg/kg) reversed the behavioral deficits, reduced proinflammatory microglia in the mPFC, normalized prefrontal Dat and Drd2 gene expression, and increased Bdnf and Nrf2 mRNA levels. These findings unravel the mechanisms underlying the beneficial effects of MPH on cognitive deficits and depressive-like behaviors observed in Mbnl2 KO mice, and suggest that MPH could be a potential candidate to treat the CNS deficiencies in DM1 patients.
Collapse
Affiliation(s)
- Carla Ramon-Duaso
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Thomas Gener
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Marta Consegal
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Cristina Fernández-Avilés
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Juan José Gallego
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Laura Castarlenas
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology and the Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.,CIBER de la Fisiopatología de la Obesidad y la Nutrición (CIBERON), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Maldonado
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.,Laboratory of Neuropharmacology, Department of Experimental al Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - M Victoria Puig
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.,Laboratory of Neuropharmacology, Department of Experimental al Health Sciences, Pompeu Fabra University, Barcelona, Spain
| |
Collapse
|
15
|
Lamanna J, Isotti F, Ferro M, Racchetti G, Anchora L, Rucco D, Malgaroli A. Facilitation of dopamine-dependent long-term potentiation in the medial prefrontal cortex of male rats follows the behavioral effects of stress. J Neurosci Res 2020; 99:662-678. [PMID: 32954528 DOI: 10.1002/jnr.24732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
The effect of stress on animal behavior and brain activity has been attracting growing attention in the last decades. Stress dramatically affects several aspects of animal behavior, including motivation and cognitive functioning, and has been used to model human pathologies such as post-traumatic stress disorder. A key question is whether stress alters the plastic potential of synaptic circuits. In this work, we evaluated if stress affects dopamine (DA)-dependent synaptic plasticity in the medial prefrontal cortex (mPFC). On male adolescent rats, we characterized anxiety- and depressive-like behaviors using behavioral testing before and after exposure to a mild stress (elevated platform, EP). After the behavioral protocols, we investigated DA-dependent long-term potentiation (DA-LTP) and depression (DA-LTD) on acute slices of mPFC and evaluated the activation of DA-producing brain regions by western and dot blot analysis. We show that exposure to the EP stress enhances DA-LTP and that desipramine (DMI) treatment abolishes this effect. We also found that DA-LTD is not affected by EP stress unless when this is followed by DMI treatment. In addition, EP stress reduces anxiety, an effect abolished by both DMI and ketamine, while motivation is promoted by previous exposure to EP stress independently of pharmacological treatments. Finally, this form of stress reduces the expression of the early gene cFOS in the ventral tegmental area. These findings support the idea that mild stressors can promote synaptic plasticity in PFC through a dopaminergic mechanism, an effect that might increase the sensitivity of mPFC to subsequent stressful experiences.
Collapse
Affiliation(s)
- Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy.,Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Isotti
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy.,Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy.,Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Gabriella Racchetti
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy.,Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Lavinia Anchora
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Daniele Rucco
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy.,Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
16
|
Developmental effects of environmental enrichment on selective and auditory sustained attention. Psychoneuroendocrinology 2020; 111:104479. [PMID: 31704636 DOI: 10.1016/j.psyneuen.2019.104479] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/23/2019] [Accepted: 10/11/2019] [Indexed: 01/29/2023]
Abstract
Environmental enrichment (EE) has been used as a positive manipulation in different disease models. However, there is conflicting evidence reported in the literature about the effects of EE. Additionally, the time period that would be most beneficial in implementing environmental enrichment as an intervention is not clear. Our study aimed to systematically compare the prenatal, juvenile, mid-adolescence, and adulthood developmental trajectory to further the understanding of enriched environment's effects on selective and auditory sustained attention, corresponding to behavioral (conceived) and physiological-reflexive (non-conceived) measures. Rats were exposed for 21 days to enriched environment during various developmental periods and compared to age-matched controls. All groups were tested for long-term effects (at postnatal day 120 and onward) on selective and sustained attention. We found that the exposure to enriched environment during mid-adolescence has yielded the most significant and long-term pattern of effects, including selective and auditory sustained attention performance, increased foraging-like behavior and a significant decrease in corticosterone level. Similarly, the exposure to EE at juvenile period improved selective attention, increased foraging-like behavior, and reduced anxiety levels as reflected in the open field as well as in low corticosterone levels. These results specify a crucial period along the developmental trajectory for applying environmental enrichment. Mid-adolescence is suggested, in future basic and translational studies, as the sensitive time period that induces the most beneficial and long-term effects of EE on attention. The current findings suggest that the exposure to EE during mid-adolescence should be further considered and studied as behavioral alternative intervention, or as adjuvant behavioral therapy, aimed to decrease the probability to develop ADHD in post-adolescence period. This suggestion is highly relevant due to the debate regarding the pros and cons of screens usage (e.g. Facebook, online games, etc.) during early life that decreases environmental enrichment, especially, direct social interaction.
Collapse
|
17
|
Waheed A, Dalton B, Wesemann U, Ibrahim MAA, Himmerich H. A Systematic Review of Interleukin-1β in Post-Traumatic Stress Disorder: Evidence from Human and Animal Studies. J Interferon Cytokine Res 2019; 38:1-11. [PMID: 29328883 DOI: 10.1089/jir.2017.0088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pro-inflammatory cytokines, such as interleukin (IL)-1β, have been implicated as underlying pathophysiological mechanisms and potential biomarkers of post-traumatic stress disorder (PTSD). This systematic review examines data regarding IL-1β production/concentration in human and animal studies of PTSD. In accordance with PRISMA guidelines, relevant articles from PubMed were reviewed from inception until July 10, 2017. Nineteen studies were eligible for inclusion. Animal studies demonstrated increased hippocampal IL-1β in rodent models of PTSD. Several immunomodulatory drugs were shown to reduce elevated IL-1β levels and anxiety-like behaviors in animals. Human cross-sectional studies showed contradictory results; serum and plasma IL-1β concentrations in PTSD patients were either elevated or did not differ from control groups. In vitro IL-1β production by stimulated cells demonstrated no difference between PTSD and control participants, although spontaneous in vitro production of IL-1β was increased in the PTSD group. The findings from 2 longitudinal studies were inconsistent. Given the conflicting findings, it is premature to consider IL-1β as a biomarker of PTSD. Anti-inflammatory agents may reduce IL-1β, and be a potential basis for future therapeutic agents in PTSD treatment. More longitudinal research is needed to better understand the role of IL-1β in the development and/or maintenance of PTSD.
Collapse
Affiliation(s)
- Aysha Waheed
- 1 Department of Psychological Medicine, King's College London , London, United Kingdom .,2 Faculty of Life Sciences and Medicine, King's College London , London, United Kingdom
| | - Bethan Dalton
- 1 Department of Psychological Medicine, King's College London , London, United Kingdom
| | - Ulrich Wesemann
- 3 Department of Psychiatry, Psychotherapy and Psychotraumatology, Bundeswehr Hospital , Berlin, Germany
| | - Mohammad A A Ibrahim
- 4 Department of Immunological Medicine and Allergy, King's Health Partners, King's College Hospital , London, United Kingdom
| | - Hubertus Himmerich
- 1 Department of Psychological Medicine, King's College London , London, United Kingdom
| |
Collapse
|
18
|
Zubedat S, Havkin E, Maoz I, Aga-Mizrachi S, Avital A. A probabilistic model of startle response reveals opposite effects of acute versus chronic Methylphenidate treatment. J Neurosci Methods 2019; 327:108389. [PMID: 31415846 DOI: 10.1016/j.jneumeth.2019.108389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/30/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND The startle response is considered as the major physio-behavioral indication of anxiety in health and disease conditions. However, due to different protocols of stimulation and measurement, the magnitude as well as the appearance of the startle response is inconsistent. NEW METHOD We postulate that the startle probability and not merely the amplitude may bare information that will form a consistent physiological measure of anxiety. RESULTS To examine the proof-of-concept of our suggested probability model, we evaluated the effects of acute (single) versus chronic (14 days) MPH administration on both startle amplitude and probability. We found that both acute and chronic MPH administration has yielded similar effects on startle amplitude. However, acute MPH increased the startle's probability while chronic MPH decreased it. Next, we evaluated the effects of acute versus chronic stress on the startle's parameters and found a complementary effect. Explicitly, acute stress increased the startle's probability while chronic stress increased the startle amplitude. In contrast, enriched environment had no significant effects. Finally, to further validate the probability measure, we show that Midazolam had significant anxiolytic effects. In the second part, we investigated the acoustic startle response parameters (e.g. background noise and pulse duration), to better understand the interplay between these parameters and the startle amplitude versus probability. CONCLUSIONS We show that the probabilistic element of the startle response does not only point to deeper physiologic relationships but may also serve as "hidden variables" congruent but not entirely identical to the commonly researched amplitude of the startle response.
Collapse
Affiliation(s)
- Salman Zubedat
- Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Evgeny Havkin
- Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inon Maoz
- Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shlomit Aga-Mizrachi
- Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avi Avital
- Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
19
|
Deslauriers J, Toth M, Zhou X, Risbrough VB. Heritable Differences in Catecholamine Signaling Modulate Susceptibility to Trauma and Response to Methylphenidate Treatment: Relevance for PTSD. Front Behav Neurosci 2019; 13:111. [PMID: 31164811 PMCID: PMC6534065 DOI: 10.3389/fnbeh.2019.00111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/02/2019] [Indexed: 12/31/2022] Open
Abstract
Alterations in cortical catecholamine signaling pathways can modulate acute and enduring responses to trauma. Heritable variation in catecholamine signaling is produced by a common functional polymorphism in the catechol-O-methyltransferase (COMT), with Val carriers exhibiting greater degradation of catecholamines than Met carriers. Furthermore, it has recently been suggested that drugs enhancing cortical catecholamine signaling may be a new therapeutic approach for posttraumatic stress disorder (PTSD) patients. We hypothesized that heritable differences in catecholamine signaling regulate the behavioral response to trauma, and that methylphenidate (MPD), a drug that preferentially blocks catecholamine reuptake in the prefrontal cortex (PFC), exerts COMT-dependent effects on trauma-induced behaviors. We first examined the contribution of the functional mutation COMTval158met to modulate enduring behavioral responses to predator stress in a unique "humanized" COMTval158met mouse line. Animals were exposed to a predator (cat) for 10 min and enduring avoidance behaviors were examined in the open field, light-dark box, and "trauma-reminder" tests 1-2 weeks later. Second, we examined the efficacy of chronic methylphenidate to reverse predator stress effects and if these effects were modulated by COMTval158met genotype. Mice were exposed to predator stress and began treatment with either saline or methylphenidate (3 mg/kg/day) 1 week after stress until the end of the testing [avoidance behaviors, working memory, and social preference (SP)]. In males, predator stress and COMTval158met had an additive effect on enduring anxiety-like behavior, with Val stressed mice showing the strongest avoidance behavior after stress compared to Met carriers. No effect of COMT genotype was observed in females. Therefore methylphenidate effects were investigated only in males. Chronic methylphenidate treatment reversed the stress-induced avoidance behavior and increased social investigation independently of genotype. Methylphenidate effects on working memory, however, were genotype-dependent, decreasing working memory in non-stressed Met carriers, and improving stress-induced working memory deficit in Val carriers. These results suggest that heritable variance in catecholamine signaling modulates the avoidance response to an acute trauma. This work supports recent human findings that methylphenidate might be a therapeutic alternative for PTSD patients and suggests that methylphenidate effects on anxiety (generalized avoidance, social withdrawal) vs. cognitive (working memory) symptoms may be modulated through COMT-independent and dependent mechanisms, respectively.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, United States
| | - Mate Toth
- Department of Behavioural Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Xianjin Zhou
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, United States.,Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
20
|
Malikowska-Racia N, Sałat K, Nowaczyk A, Fijałkowski Ł, Popik P. Dopamine D2/D3 receptor agonists attenuate PTSD-like symptoms in mice exposed to single prolonged stress. Neuropharmacology 2019; 155:1-9. [PMID: 31085186 DOI: 10.1016/j.neuropharm.2019.05.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 04/28/2019] [Accepted: 05/09/2019] [Indexed: 01/19/2023]
Abstract
Medications that enhance dopaminergic neurotransmission can be useful in the pharmacotherapy of posttraumatic stress disorder (PTSD), which manifests as fearful memory retrieval, anxiety and depression. We examined the effects of subchronic (15 days) treatment with select dopaminergic medications, including bromocriptine, modafinil, dihydrexidine, rotigotine and pramipexole, in a mouse model of PTSD induced by single prolonged stress (mSPS). The potential antidepressant-like and anxiolytic effects of the medications were measured by the forced swim test (FST) and the elevated plus maze (EPM) test, respectively. In addition, we studied the effects of these medications on memory retrieval in an auditory fear conditioning (FC) test, on ultrasonic vocalizations (USVs) induced by restraint stress, and on spontaneous locomotor activity (SLA). We report that a single exposure to a severe and complex set of stressors several days before testing increased immobility time in the FST and freezing in the FC paradigm and reduced the time spent in the open arms of the EPM. The stressed mice also displayed increased USVs, especially the short type. While none of the tested dopamine-mimetics exhibited anxiolytic-like effects, rotigotine produced antidepressant-like activity specifically in the mSPS-exposed animals. Moreover, both rotigotine and pramipexole shortened the duration of freezing in the fear conditioning test, but only in the mSPS-exposed mice. This study supports the hypothesis that the activation of dopaminergic D2/D3 receptors may be a promising pharmacotherapy for PTSD.
Collapse
Affiliation(s)
- Natalia Malikowska-Racia
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland.
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094, Bydgoszcz, Poland
| | - Łukasz Fijałkowski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094, Bydgoszcz, Poland
| | - Piotr Popik
- Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michalowskiego St., 31-126, Krakow, Poland; Behavioral Neuroscience and Drug Development, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343, Krakow, Poland
| |
Collapse
|
21
|
Jager A, Kanters D, Geers F, Buitelaar JK, Kozicz T, Glennon JC. Methylphenidate Dose-Dependently Affects Aggression and Improves Fear Extinction and Anxiety in BALB/cJ Mice. Front Psychiatry 2019; 10:768. [PMID: 31708820 PMCID: PMC6823535 DOI: 10.3389/fpsyt.2019.00768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022] Open
Abstract
Overt aggression, increased anxiety, and dysfunctional fear processing are often observed in individuals with conduct disorder (CD) and attention-deficit hyperactivity disorder (ADHD). Methylphenidate (MPH), a psychostimulant increasing dopamine and noradrenaline tone, is effective in reducing aggression in both CD and ADHD individuals. However, it is unclear to which extent these effects of MPH are dose dependent. Here, the effects of acute intraperitoneal MPH (3 and 10 mg/kg) on aggression, anxiety, social behavior, and fear extinction were investigated in BALB/cJ mice. Previous studies in BALB/cJ mice have revealed high levels of aggression and anxiety that are associated with reduced top-down cortical control. Administration of 3 mg/kg MPH prolonged the attack latency and prevented escalation of aggression over time compared to vehicle-treated mice, while 10 mg/kg MPH increased number of bites and attacks. In addition, 3 mg/kg MPH decreased social interaction slightly. A strong anxiolytic effect was found after administration of both the 3 and 10 mg/kg doses in the elevated plus maze and the open-field test. In addition, while vehicle-treated BALB/cJ animals showed intact freezing, both doses of MPH decreased freezing to the unconditioned stimulus in a fear-conditioning paradigm. A long-lasting effect on fear extinction was visible after treatment with the 10 mg/kg dose. The data support a role for MPH in the regulation of anxiety, fear processing, and aggression in BALB/cJ mice, with the latter effect in a dose-dependent manner. The findings provide a further context for examining the effects of MPH in clinical disorders such as ADHD and CD.
Collapse
Affiliation(s)
- Amanda Jager
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Doranda Kanters
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Femke Geers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands.,Department of Clinical Genomics, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
22
|
Hsu SPC, Wang DY, Min MY, Fu YS. Long-term challenge of methylphenidate changes the neuronal population and membrane property of dopaminergic neuron in rats. Neurochem Int 2019; 122:187-195. [DOI: 10.1016/j.neuint.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/28/2018] [Accepted: 11/02/2018] [Indexed: 01/08/2023]
|
23
|
Howlett JR, Campbell-Sills L, Jain S, Heeringa SG, Nock MK, Sun X, Ursano RJ, Stein MB. Attention Deficit Hyperactivity Disorder and Risk of Posttraumatic Stress and Related Disorders: A Prospective Longitudinal Evaluation in U.S. Army Soldiers. J Trauma Stress 2018; 31:909-918. [PMID: 30461069 PMCID: PMC6386189 DOI: 10.1002/jts.22347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 07/01/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
Cross-sectional associations between attention deficit hyperactivity disorder (ADHD) and posttraumatic stress disorder (PTSD) have been observed, but longitudinal studies assessing this association are lacking. This prospective study evaluated the association between predeployment ADHD and postdeployment PTSD among U.S. Army soldiers. Soldiers who deployed to Afghanistan were surveyed before deployment (T0) and approximately 1 month (T1), 3 months (T2), and 9 months (T3) after their return. Logistic regression was performed to estimate the association between predeployment ADHD and postdeployment (T2 or T3) PTSD among 4,612 soldiers with data at all waves and no record of stimulant medication treatment during the study. To evaluate specificity of the ADHD-PTSD association, we examined associations among predeployment ADHD, postdeployment major depressive episode (MDE), generalized anxiety disorder (GAD), and suicidal ideation. Weighted prevalence of ADHD predeployment was 6.1% (SE = 0.4%). Adjusting for other risk factors, predeployment ADHD was associated with risk of postdeployment PTSD, adjusted odds ratio (AOR) = 2.13, 95% CI [1.51, 3.00], p < .001, including incidence among soldiers with no predeployment history of PTSD, AOR = 2.50, 95% CI [1.69, 3.69], p < .001. ADHD was associated with postdeployment MDE, AOR = 2.80, 95% CI [2.01, 3.91], p < .001, and GAD, AOR = 3.04, 95% CI [2.10, 4.42], p < .001, but not suicidal ideation. Recognition of associations between predeployment ADHD and postdeployment PTSD, MDE, and GAD may inform targeted prevention efforts. Future research should examine whether treatment of ADHD is protective against PTSD and related disorders in trauma-exposed individuals.
Collapse
Affiliation(s)
- Jonathon R. Howlett
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Laura Campbell-Sills
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Sonia Jain
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Steven G. Heeringa
- University of Michigan, Institute for Social Research, Ann Arbor, Michigan, USA
| | - Matthew K. Nock
- Department of Psychology, Harvard University, Cambridge, Massachusetts, USA
| | - Xiaoying Sun
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Robert J. Ursano
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Murray B. Stein
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA,Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA,VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
24
|
Felger JC. Imaging the Role of Inflammation in Mood and Anxiety-related Disorders. Curr Neuropharmacol 2018; 16:533-558. [PMID: 29173175 PMCID: PMC5997866 DOI: 10.2174/1570159x15666171123201142] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 10/23/2017] [Accepted: 11/21/2017] [Indexed: 02/08/2023] Open
Abstract
Background Studies investigating the impact of a variety of inflammatory stimuli on the brain and behavior have reported evidence that inflammation and release of inflammatory cytokines affect circuitry relevant to both reward and threat sensitivity to contribute to behavioral change. Of relevance to mood and anxiety-related disorders, biomarkers of inflammation such as inflammatory cytokines and acute-phase proteins are reliably elevated in a significant proportion of patients with major depressive disorder (MDD), bipolar disorder, anxiety disorders and post-traumatic stress disorder (PTSD). Methods This review summarized clinical and translational work demonstrating the impact of peripheral inflammation on brain regions and neurotransmitter systems relevant to both reward and threat sensitivity, with a focus on neuroimaging studies involving administration of inflammatory stimuli. Recent translation of these findings to further understand the role of inflammation in mood and anxiety-related disorders is also discussed. Results Inflammation was consistently found to affect basal ganglia and cortical reward and motor circuits to drive reduced motivation and motor activity, as well as anxiety-related brain regions including amygdala, insula and anterior cingulate cortex, which may result from cytokine effects on monoamines and glutamate. Similar relationships between inflammation and altered neurocircuitry have been observed in MDD patients with increased peripheral inflammatory markers, and such work is on the horizon for anxiety disorders and PTSD. Conclusion Neuroimaging effects of inflammation on reward and threat circuitry may be used as biomarkers of inflammation for future development of novel therapeutic strategies to better treat mood and anxiety-related disorders in patients with high inflammation.
Collapse
Affiliation(s)
- Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States.,Winship Cancer Institute, Emory University, Atlanta, GA, United States
| |
Collapse
|
25
|
Oh JY, Kim YK, Kim SN, Lee B, Jang JH, Kwon S, Park HJ. Acupuncture modulates stress response by the mTOR signaling pathway in a rat post-traumatic stress disorder model. Sci Rep 2018; 8:11864. [PMID: 30089868 PMCID: PMC6082850 DOI: 10.1038/s41598-018-30337-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 07/25/2018] [Indexed: 01/02/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disease that can form following exposure to a traumatic event. Acupuncture has been proposed as a beneficial treatment for PTSD, but the underlying mechanisms remain unclear. The present study investigated whether acupuncture improves depression- and anxiety-like behaviors induced using a single prolonged stress (SPS) as a PTSD rat model. In addition, we investigated whether the effects were mediated by increased mTOR activity and its downstream signaling components, which contribute to protein synthesis required for synaptic plasticity in the hippocampus. We found that acupuncture at HT8 significantly alleviated both depression- and anxiety-like behaviors induced by SPS in rats, as assessed by the forced swimming, elevated plus maze, and open field tests; this alleviation was blocked by rapamycin. The effects of acupuncture were equivalent to those exerted by fluoxetine. Acupuncture regulated protein translation in the mTOR signaling pathway and enhanced the activation of synaptic proteins, PSD95, Syn1, and GluR1 in the hippocampus. These results suggest that acupuncture exerts antidepressant and anxiolytic effects on PTSD-related symptoms by increasing protein synthesis required for synaptic plasticity via the mTOR pathway in the hippocampus. Acupuncture may be a promising treatment for patients with PTSD and play a role as an alternative PTSD treatment.
Collapse
Affiliation(s)
- Ju-Young Oh
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Yu-Kang Kim
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Seung-Nam Kim
- College of Korean Medicine, Dongguk University, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Jae-Hwan Jang
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea
| | - Sunoh Kwon
- Korean Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea. .,Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea. .,BK21 PLUS Korean Medicine Science Center, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemoon-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
26
|
Coelho-Santos V, Cardoso FL, Leitão RA, Fontes-Ribeiro CA, Silva AP. Impact of developmental exposure to methylphenidate on rat brain's immune privilege and behavior: Control versus ADHD model. Brain Behav Immun 2018; 68:169-182. [PMID: 29061363 DOI: 10.1016/j.bbi.2017.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/19/2017] [Accepted: 10/19/2017] [Indexed: 12/15/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is the most prevalent childhood mental disorders that often persists into adulthood. Moreover, methylphenidate (MPH) is the mainstay of medical treatment for this disorder. Yet, not much is known about the neurobiological impact of MPH on control versus ADHD conditions, which is crucial to simultaneously clarify the misuse/abuse versus therapeutic use of this psychostimulant. In the present study, we applied biochemical and behavioral approaches to broadly explore the early-life chronic exposure of two different doses of MPH (1.5 and 5 mg/kg/day) on control and ADHD rats (Wistar Kyoto and Spontaneously Hypertensive rats, respectively). We concluded that the higher dose of MPH promoted blood-brain barrier (BBB) permeability and elicited anxiety-like behavior in both control and ADHD animals. BBB dysfunction triggered by MPH was particularly prominent in control rats, which was characterized by a marked disruption of intercellular junctions, an increase of endothelial vesicles, and an upregulation of adhesion molecules concomitantly with the infiltration of peripheral immune cells into the prefrontal cortex. Moreover, both doses of MPH induced a robust neuroinflammatory and oxidative response in control rats. Curiously, in the ADHD model, the lower dose of MPH (1.5 mg/kg/day) had a beneficial effect since it balanced both immunity and behavior relative to vehicle animals. Overall, the contrasting effects of MPH observed between control and ADHD models support the importance of an appropriate MPH dose regimen for ADHD, and also suggest that MPH misuse negatively affects brain and behavior.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Filipa L Cardoso
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ricardo A Leitão
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Carlos A Fontes-Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
27
|
Bou Khalil R, Smayra V, Saliba Y, Hajal J, Bakhos JJ, Souaiby L, Richa S, Tamraz J, Farès N. Mifepristone reduces hypothalamo-pituitary-adrenal axis activation and restores weight loss in rats subjected to dietary restriction and methylphenidate administration. Neurosci Res 2017; 135:46-53. [PMID: 29288690 DOI: 10.1016/j.neures.2017.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/16/2017] [Accepted: 12/22/2017] [Indexed: 11/16/2022]
Abstract
This study evaluates the efficacy of mifepristone on weight restoration in rats subjected to dietary restriction and methylphenidate administration. 25 female rats aged between 9 and 12 months were divided into 2 groups: 5 controls (exposed only to dietary restriction) and 20 rats that were administered 5 mg/kg/d of methylphenidate before meal exposure, for 36 days. Among rats who responded to methylphenidate (weight loss of 15-25%) weeks after its administration, a group of 6 rats continued to receive only methylphenidate ("Met" group), and another group received 10 mg/kg/d of mifepristone in addition to methylphenidate for 18 days ("Met+Mif" group; n = 6). The mean weight of the "Met+Mif" group remained significantly lower when compared to the control group (87.63 ± 2.83% vs 96.29 ± 3.26%; p < 0.001 respectively) but was significantly higher than that of the "Met" group (87.63 ± 2.83% vs. 80.61 ± 3.52%; p < 0.001 respectively). Plasma concentrations of adiponectin and gene expression of its receptors in rats brain were significantly higher in the "Met" group as compared to the "Met+Mif" and control groups (p < 0.01). Accordingly, mifepristone reduces HPA axis activation and restores weight through adipose tissue recovering. It might be considered a promising treatment for anorexia nervosa patients in future studies.
Collapse
Affiliation(s)
- Rami Bou Khalil
- Saint Joseph University, Hôtel Dieu de France Hospital, Department of Psychiatry, Beirut, Lebanon; Saint Joseph University, Research Laboratory in Physiology and Physiopathology, LRPP, Beirut, Lebanon; Saint Joseph University, Faculty of Medicine, Beirut, Lebanon.
| | - Viviane Smayra
- Saint Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Youakim Saliba
- Saint Joseph University, Research Laboratory in Physiology and Physiopathology, LRPP, Beirut, Lebanon
| | - Joelle Hajal
- Saint Joseph University, Research Laboratory in Physiology and Physiopathology, LRPP, Beirut, Lebanon
| | - Jules-Joël Bakhos
- Saint Joseph University, Research Laboratory in Physiology and Physiopathology, LRPP, Beirut, Lebanon
| | - Lama Souaiby
- National mental health program, Ministry of public health, Beirut, Lebanon
| | - Sami Richa
- Saint Joseph University, Hôtel Dieu de France Hospital, Department of Psychiatry, Beirut, Lebanon; Saint Joseph University, Faculty of Medicine, Beirut, Lebanon; Saint Joseph University, Head of department of Psychiatry, Beirut, Lebanon
| | - Jean Tamraz
- Saint Joseph University, Faculty of Medicine, Beirut, Lebanon; Saint Joseph University, Hôtel Dieu de France Hospital, Department of Neuroimaging, Beirut, Lebanon
| | - Nassim Farès
- Saint Joseph University, Research Laboratory in Physiology and Physiopathology, LRPP, Beirut, Lebanon; Saint Joseph University, Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
28
|
Schreiber S, Bader M, Rubovitch V, Pick CG. Interaction between methylphenidate, methadone and different antidepressant drugs on antinociception in mice, and possible clinical implications. World J Biol Psychiatry 2017; 18:300-307. [PMID: 26529542 DOI: 10.3109/15622975.2015.1086492] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Methylphenidate (MPH), a psychostimulant used for treatment of attention deficit hyperactivity disorder (ADHD), is widely used by patients on antidepressants and methadone maintenance treatment (MMT). Preclinical studies showed MPH to exert analgesic effects when given alone or with morphine. METHODS Using the hotplate assay on mice, we studied the interaction of acute doses of MPH with sub-threshold doses of methadone and different antidepressant medications and the interaction of increasing doses of MPH with chronic methadone. RESULTS Adding a sub-threshold dose of venlafaxine, desipramine or clomipramine to MPH produced significant augmentation of MPH antinociception with each medication (P < 0.05). No such interactions were found between escitalopram and acute methadone. However, addition of increasing doses of MPH to chronic methadone given for 2 weeks using ALZET osmotic mini pumps induced augmentation of the antinociceptive effect of chronic methadone exclusively at high dose of MPH (7.5 mg/kg). CONCLUSIONS These findings may implicate the need of an excessive attention to the administration of MPH to MMT patients. The no interaction found between MPH and escitalopram may hint to the possibly safe co-administration of MPH and selective serotonin reuptake inhibitors (SSRIs) to depressed ADHD patients. Further studies are needed in order to validate these possible clinical implications.
Collapse
Affiliation(s)
- Shaul Schreiber
- a Department of Psychiatry , Tel Aviv Sourasky Medical Center & Tel Aviv University Sackler Faculty of Medicine , Tel Aviv , Israel
| | - Miaad Bader
- b Department of Anatomy, and Anthropology , Sackler Faculty of Medicine
| | - Vardit Rubovitch
- b Department of Anatomy, and Anthropology , Sackler Faculty of Medicine
| | - Chaim G Pick
- b Department of Anatomy, and Anthropology , Sackler Faculty of Medicine.,c Sagol School of Neuroscience, Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
29
|
Deslauriers J, Powell S, Risbrough VB. Immune signaling mechanisms of PTSD risk and symptom development: insights from animal models. Curr Opin Behav Sci 2017; 14:123-132. [PMID: 28758144 DOI: 10.1016/j.cobeha.2017.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized by persistent re-experiencing of a traumatic event, avoidance, and increased arousal. The approved pharmacological treatments for PTSD have limited efficacy (~60% treatment response), supporting the need for identification of biomarkers and novel pharmacological therapies. Mounting evidence suggests increased inflammatory markers and altered immune gene expression correlate with the severity of symptoms in PTSD patients. However a causal role of immune signaling in development and maintenance of PTSD symptoms is not clear, as inflammation may also be an epiphenomenon related to metabolic and behavioral effects of stress. Animal studies have been critical in understanding the potential causal role of immune signaling in PTSD. In this review we will present the most recent evidence, primarily focusing on the last 3 years, for inflammatory dysfunction both preceding and following PTSD, and how animal models of PTSD have contributed to our understanding of immune mechanisms involved in enduring anxiety after trauma. We will particularly focus on the role of peripheral vs. central immune signaling, the differences between single vs. chronic stress models of PTSD and recent utilization of these models to investigate novel anti-inflammatory treatments. We also highlight some current gaps in the literature including models of TBI/PTSD comorbidity, lack of translational peripheral markers of inflammation and the relatively incomplete understanding of the inflammatory trajectory after severe stress.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Susan Powell
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| |
Collapse
|
30
|
Herbst ED, McCaslin SE, Kalapatapu RK. Use of Stimulants and Performance Enhancers During and After Trauma Exposure in a Combat Veteran: A Possible Risk Factor for Posttraumatic Stress Symptoms. Am J Psychiatry 2017; 174:95-99. [PMID: 28142270 PMCID: PMC5289819 DOI: 10.1176/appi.ajp.2016.16010014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ellen D. Herbst
- Mental Health Services, San Francisco VA Health Care System (SFVAHCS),Department of Psychiatry, University of California, San Francisco (UCSF)
| | - Shannon E. McCaslin
- National Center for PTSD, VA Palo Alto Health Care System, Dissemination & Training Division
| | - Raj K. Kalapatapu
- Mental Health Services, San Francisco VA Health Care System (SFVAHCS),Department of Psychiatry, University of California, San Francisco (UCSF)
| |
Collapse
|
31
|
Ritov G, Richter-Levin G. Pre-trauma Methylphenidate in rats reduces PTSD-like reactions one month later. Transl Psychiatry 2017; 7:e1000. [PMID: 28072410 PMCID: PMC5545737 DOI: 10.1038/tp.2016.277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/01/2016] [Accepted: 11/27/2016] [Indexed: 01/10/2023] Open
Abstract
In basic research, the etiology of fear-related pathologies, such as post-traumatic stress disorder (PTSD), is conceptualized using fear-conditioning protocols that pair environmental stimuli (that is, a conditioned stimulus-CS) with an aversive, unconditioned stimulus (US) to elicit an assessable conditioned fear response. Although pathophysiological models agree that regulatory dysfunctions in this associative process may instigate fear-related pathology, current opinions differ in regard to the nature of these dysfunctions. Primarily derived from studies in rodents, the prevailing perspective proposes that pathological fear-reactions develop from intensified and overly consolidated CS-US associations. Alternatively, models derived from studies in humans suggest that tempospatial inaccuracies in representations of associative fear might precipitate pathology by engendering failure to differentiate present experiences and past memories of threat. To test this concept in rodents, we administered rats with cognition enhancing doses of Methylphenidate before or after fear conditioning and measured long-term alterations in their conditioned fear behaviors and PTSD-like reactions. The administration of Methylphenidate before fear-memory formation indeed reduced anxious-like responses during fear-memory retrieval one month later. An individual profiling analysis revealed that Methylphenidate onset had opposing effects on the risk for PTSD-like classification. The modulation of initial learning and formation of associative fear normalized the risk for developing PTSD-like reaction. In contrast, when the effects of Methylphenidate were exerted only over later consolidation this risk increased markedly. When examined under current psychiatric and neuropharmacologic literature, these results reveal a possible strategy of using low-dose Methylphenidate for the prevention of PTSD in high risk populations.
Collapse
Affiliation(s)
- G Ritov
- The Institute for the Study of Affective Neuroscience, University of Haifa, Haifa, Israel,Sagol Department of Neurobiology, University of Haifa, Haifa, Israel,Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel,Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel E-mail:
| | - G Richter-Levin
- The Institute for the Study of Affective Neuroscience, University of Haifa, Haifa, Israel,Sagol Department of Neurobiology, University of Haifa, Haifa, Israel,Psychology Department, University of Haifa, Haifa, Israel,Sagol Department of Neurobiology, University of Haifa, Haifa 31905, Israel. E-mail:
| |
Collapse
|
32
|
Randomized Placebo-Controlled Trial of Methylphenidate or Galantamine for Persistent Emotional and Cognitive Symptoms Associated with PTSD and/or Traumatic Brain Injury. Neuropsychopharmacology 2016; 41:1191-8. [PMID: 26361060 PMCID: PMC4793116 DOI: 10.1038/npp.2015.282] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 01/04/2023]
Abstract
We report findings from a 12-week randomized double-blinded placebo-controlled trial of methylphenidate or galantamine to treat emotional and cognitive complaints in individuals (n=32) with a history of PTSD, TBI, or both conditions. In this small pilot study, methylphenidate treatment was associated with clinically meaningful and statistically significant improvement compared with placebo on the primary outcome, a measure of cognitive complaints (Ruff Neurobehavioral Inventory-Postmorbid Cognitive Scale), as well as on the secondary outcomes reflecting post-concussive (Rivermead Post Concussive Symptom Questionnaire) and post-traumatic stress symptoms (Posttraumatic Stress Disorder Checklist). Treatment was well tolerated. These results suggest the need for a larger RCT to replicate and confirm these findings. Design considerations for such a trial should include the need for multiple sites to facilitate adequate recruitment and extension of the treatment and follow-up periods.
Collapse
|
33
|
cis-3-Hexenol and trans-2-hexenal mixture prevents development of PTSD-like phenotype in rats. Behav Brain Res 2016; 297:251-8. [DOI: 10.1016/j.bbr.2015.10.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 10/03/2015] [Accepted: 10/08/2015] [Indexed: 02/05/2023]
|
34
|
Vainshtein A, Veenman L, Shterenberg A, Singh S, Masarwa A, Dutta B, Island B, Tsoglin E, Levin E, Leschiner S, Maniv I, Pe’er L, Otradnov I, Zubedat S, Aga-Mizrachi S, Weizman A, Avital A, Marek I, Gavish M. Quinazoline-based tricyclic compounds that regulate programmed cell death, induce neuronal differentiation, and are curative in animal models for excitotoxicity and hereditary brain disease. Cell Death Discov 2015; 1:15027. [PMID: 27551459 PMCID: PMC4979516 DOI: 10.1038/cddiscovery.2015.27] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 12/21/2022] Open
Abstract
Expanding on a quinazoline scaffold, we developed tricyclic compounds with biological activity. These compounds bind to the 18 kDa translocator protein (TSPO) and protect U118MG (glioblastoma cell line of glial origin) cells from glutamate-induced cell death. Fascinating, they can induce neuronal differentiation of PC12 cells (cell line of pheochromocytoma origin with neuronal characteristics) known to display neuronal characteristics, including outgrowth of neurites, tubulin expression, and NeuN (antigen known as 'neuronal nuclei', also known as Rbfox3) expression. As part of the neurodifferentiation process, they can amplify cell death induced by glutamate. Interestingly, the compound 2-phenylquinazolin-4-yl dimethylcarbamate (MGV-1) can induce expansive neurite sprouting on its own and also in synergy with nerve growth factor and with glutamate. Glycine is not required, indicating that N-methyl-D-aspartate receptors are not involved in this activity. These diverse effects on cells of glial origin and on cells with neuronal characteristics induced in culture by this one compound, MGV-1, as reported in this article, mimic the diverse events that take place during embryonic development of the brain (maintenance of glial integrity, differentiation of progenitor cells to mature neurons, and weeding out of non-differentiating progenitor cells). Such mechanisms are also important for protective, curative, and restorative processes that occur during and after brain injury and brain disease. Indeed, we found in a rat model of systemic kainic acid injection that MGV-1 can prevent seizures, counteract the process of ongoing brain damage, including edema, and restore behavior defects to normal patterns. Furthermore, in the R6-2 (transgenic mouse model for Huntington disease; Strain name: B6CBA-Tg(HDexon1)62Gpb/3J) transgenic mouse model for Huntington disease, derivatives of MGV-1 can increase lifespan by >20% and reduce incidence of abnormal movements. Also in vitro, these derivatives were more effective than MGV-1.
Collapse
Affiliation(s)
- A Vainshtein
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - L Veenman
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - A Shterenberg
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - S Singh
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - A Masarwa
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - B Dutta
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - B Island
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - E Tsoglin
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - E Levin
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - S Leschiner
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - I Maniv
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - L Pe’er
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - I Otradnov
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - S Zubedat
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - S Aga-Mizrachi
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - A Weizman
- Tel Aviv University, Sackler Faculty of Medicine, The Felsenstein Medical Research Center, Geha Mental Health Center, Tel Aviv, Israel
| | - A Avital
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - I Marek
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - M Gavish
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| |
Collapse
|
35
|
Safory H, Neame S, Shulman Y, Zubedat S, Radzishevsky I, Rosenberg D, Sason H, Engelender S, Avital A, Hülsmann S, Schiller J, Wolosker H. The alanine-serine-cysteine-1 (Asc-1) transporter controls glycine levels in the brain and is required for glycinergic inhibitory transmission. EMBO Rep 2015; 16:590-8. [PMID: 25755256 DOI: 10.15252/embr.201439561] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/13/2015] [Indexed: 11/09/2022] Open
Abstract
Asc-1 (SLC7A10) is an amino acid transporter whose deletion causes neurological abnormalities and early postnatal death in mice. Using metabolomics and behavioral and electrophysiological methods, we demonstrate that Asc-1 knockout mice display a marked decrease in glycine levels in the brain and spinal cord along with impairment of glycinergic inhibitory transmission, and a hyperekplexia-like phenotype that is rescued by replenishing brain glycine. Asc-1 works as a glycine and L-serine transporter, and its transport activity is required for the subsequent conversion of L-serine into glycine in vivo. Asc-1 is a novel regulator of glycine metabolism and a candidate for hyperekplexia disorders.
Collapse
Affiliation(s)
- Hazem Safory
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Samah Neame
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yoav Shulman
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Salman Zubedat
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inna Radzishevsky
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dina Rosenberg
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hagit Sason
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Simone Engelender
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avi Avital
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel Emek Medical Center, Afula, Israel
| | - Swen Hülsmann
- Department of Anesthesiology, Emergency and Intensive Care Medicine and Center for Nanoscale Microscopy and Molecular Physiology of the Brain Georg-August-University, Göttingen, Germany
| | - Jackie Schiller
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Herman Wolosker
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|