1
|
Poceviciute I, Brazaityte A, Buisas R, Vengeliene V. Scopolamine animal model of memory impairment. Behav Brain Res 2025; 479:115344. [PMID: 39566583 DOI: 10.1016/j.bbr.2024.115344] [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: 09/22/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024]
Abstract
In this study, we reassessed the suitability of a commonly used pharmacological animal model of Alzheimer's disease (AD) - scopolamine-induced memory impairment. The goal of the study was to explore if this animal model induces other behavioral changes associated with AD. One of the key behavioral features of AD, manifesting already during the early stages of the illness, is apathy-like behavior. We also evaluated how behavioral alterations induced by scopolamine compare to those seen in healthy aging animals. To achieve these goals, locomotor activity and short-term memory of young male Wistar rats were tested in the open field, novel object recognition (NOR) and T-maze spontaneous alternation tests before, during and after 21 daily administrations of scopolamine. Three-, ten- and nineteen-month-old male and female rats were used to measure age-related changes in these behaviors. Our data showed that although both scopolamine treatment and aging reduced the number of approaches to the objects and their exploration time during the NOR test, correlation with impaired object recognition memory was only observed in the scopolamine treated animals. Furthermore, treatment with scopolamine significantly increased the locomotor activity, which could be observed even one week after treatment discontinuation. Contrary, locomotor activity in older rats was significantly lower than that of younger rats. These findings demonstrate that the animal model of scopolamine-induced memory impairment fails to incorporate apathy-like symptoms characteristic to the AD and age-related reduction in physical activity of older rats.
Collapse
Affiliation(s)
- Ieva Poceviciute
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, Lithuania
| | - Agne Brazaityte
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, Lithuania
| | - Rokas Buisas
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, Lithuania
| | - Valentina Vengeliene
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, Lithuania.
| |
Collapse
|
2
|
Munarriz‐Cuezva E, Meana JJ. Poly (I:C)-induced maternal immune activation generates impairment of reversal learning performance in offspring. J Neurochem 2025; 169:e16212. [PMID: 39183542 PMCID: PMC11657921 DOI: 10.1111/jnc.16212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Maternal immune activation (MIA) induces a variety of behavioral and brain abnormalities in offspring of rodent models, compatible with neurodevelopmental disorders, such as schizophrenia or autism. However, it remains controversial whether MIA impairs reversal learning, a basic expression of cognitive flexibility that seems to be altered in schizophrenia. In the present study, MIA was induced by administration of a single dose of polyriboinosinic-polyribocytidylic acid (Poly (I:C) (5 mg/kg i.p.)) or saline to mouse pregnant dams in gestational day (GD) 9.5. Immune activation was monitored through changes in weight and temperature. The offspring were evaluated when they reached adulthood (8 weeks) using a touchscreen-based system to investigate the effects of Poly (I:C) on discrimination and reversal learning performance. After an initial pre-training, mice were trained to discriminate between two different stimuli, of which only one was rewarded (acquisition phase). When the correct response reached above 80% values for two consecutive days, the images were reversed (reversal phase) to assess the adaptation capacity to a changing environment. Maternal Poly (I:C) treatment did not interfere with the learning process but induced deficits in reversal learning compared to control saline animals. Thus, the accuracy in the reversal phase was lower, and Poly (I:C) animals required more sessions to complete it, suggesting impairments in cognitive flexibility. This study advances the knowledge of how MIA affects behavior, especially cognitive domains that are impaired in schizophrenia. The findings support the validity of the Poly (I:C)-based MIA model as a tool to develop pharmacological treatments targeting cognitive deficits associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eva Munarriz‐Cuezva
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
| | - Jose Javier Meana
- Department of Pharmacology, Faculty of Medicine and NursingUniversity of the Basque CountryLeioaBizkaiaSpain
- Centro de Investigación Biomédica en Red de Salud MentalLeioaBizkaiaSpain
- Biobizkaia Health Research InstituteBarakaldoBizkaiaSpain
| |
Collapse
|
3
|
Lieberman JA, Mendelsohn A, Goldberg TE, Emsley R. Preventing disease progression in schizophrenia: What are we waiting for. J Psychiatr Res 2024; 181:716-727. [PMID: 39754992 DOI: 10.1016/j.jpsychires.2024.12.042] [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: 09/06/2023] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Despite research advances and progress in health care, schizophrenia remains a debilitating and costly disease. Onset occurs typically during youth and can lead to a relapsing and ultimately chronic course with persistent symptoms and functional impairment if not promptly and properly treated. Consequently, over time, schizophrenia causes substantial distress and disability for patients, their families and accrues to a collective burden to society. Recent research has revealed much about the pathophysiology that underlies the progressive nature of schizophrenia. Additionally, treatment strategies for disease management have been developed that have the potential to not just control psychotic symptoms but limit the cumulative morbidity of the illness. Given the evidence for their effectiveness and feasibility for their application, it is perplexing that this model of care has not yet become the standard of care and widely implemented to reduce the burden of illness on patients and society. This begs the question of whether the failure of implementation of a potentially disease-modifying strategy is due to the lack of evidence of efficacy (or belief in it) and readiness for implementation, or whether it's the lack of motivation and political will to support their utilization. To address this question, we reviewed and summarized the literature describing the natural history, pathophysiology and therapeutic strategies that can alleviate symptoms, prevent relapse, and potentially modify the course of schizophrenia. We conclude that, while we await further advances in mental health care from research, we must fully appreciate and take advantage of the effectiveness of existing treatments and overcome the attitudinal, policy, and infrastructural barriers to providing optimal mental health care capable of providing a disease-modifying treatment to patients with schizophrenia.
Collapse
Affiliation(s)
- Jeffrey A Lieberman
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Alana Mendelsohn
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Terry E Goldberg
- Division of Geriatric Psychiatry, Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Robin Emsley
- Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
4
|
Chen Y, Gu Y, Wang B, Wei A, Dong N, Jiang Y, Liu X, Zhu L, Zhu F, Tan T, Jing Z, Mao F, Zhang Y, Yao J, Yang Y, Wang H, Wu H, Li H, Zheng C, Duan X, Huo J, Wu X, Hu S, Zhao A, Li Z, Cheng X, Qin Y, Song Q, Zhan S, Qu Q, Guan F, Xu H, Kang X, Wang C. Synaptotagmin-11 deficiency mediates schizophrenia-like behaviors in mice via dopamine over-transmission. Nat Commun 2024; 15:10571. [PMID: 39632880 PMCID: PMC11618495 DOI: 10.1038/s41467-024-54604-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Schizophrenia is a severe neuropsychiatric disease, but the initiation mechanisms are unclear. Although antipsychotics are effective against positive symptoms, therapeutic interventions for negative symptoms are limited due to the lack of pathophysiological mechanisms. Here we identify synaptotagmin-11 (Syt11) as a potential genetic risk factor and dopamine over-transmission as a mechanism in the development of schizophrenia. Syt11 expression is reduced in individuals with schizophrenia but restored following the treatment with antipsychotics. Syt11 deficiency in dopamine neurons in early adolescence, but not in adults, leads to persistent social deficits and other schizophrenia-like behaviors by mediating dopamine over-transmission in mice. Accordingly, dopamine neuron over-excitation before late adolescence induces persistent schizophrenia-associated behavioral deficits, along with the structural and functional alternations in the mPFC. Notably, local intervention of D2R with clinical drugs presynaptically or postsynaptically exhibits both acute and long-lasting therapeutic effects on social deficits in schizophrenia mice models. These findings not only define Syt11 as a risk factor and DA over-transmission as a potential risk factor initiating schizophrenia, but also propose two D2R-targeting strategies for the comprehensive and long-term recovery of schizophrenia-associated social withdrawal.
Collapse
Affiliation(s)
- Yang Chen
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhao Gu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bianbian Wang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Anqi Wei
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Nan Dong
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoying Liu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Li Zhu
- Key Laboratory of National Health Commission for Forensic Sciences, College of Medicine & Forensics, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Feng Zhu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zexin Jing
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Fenghan Mao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yichi Zhang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingyu Yao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuxin Yang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Hongyan Wang
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Hao Wu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hua Li
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Chaowen Zheng
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xueting Duan
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingxiao Huo
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xuanang Wu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shaoqin Hu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Anran Zhao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ziyang Li
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xu Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Yuhao Qin
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Qian Song
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shuqin Zhan
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qiumin Qu
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Fanglin Guan
- Key Laboratory of National Health Commission for Forensic Sciences, College of Medicine & Forensics, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Huadong Xu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Xinjiang Kang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.
| | - Changhe Wang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of Psychology, Chengwu People's Hospital, Heze, 274200, China.
| |
Collapse
|
5
|
Brand BA, de Boer JN, Willemse EJM, Weickert CS, Sommer IE, Weickert TW. Antipsychotic-induced prolactin elevation in premenopausal women with schizophrenia: associations with estrogen, disease severity and cognition. Arch Womens Ment Health 2024; 27:931-941. [PMID: 38995314 PMCID: PMC11579114 DOI: 10.1007/s00737-024-01491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
PURPOSE Antipsychotic-induced prolactin elevation may impede protective effects of estrogens in women with schizophrenia-spectrum disorders (SSD). Our study sought to confirm whether the use of prolactin-raising antipsychotics is associated with lower estrogen levels, and to investigate how estrogen and prolactin levels relate to symptom severity and cognition in premenopausal women with SSD. METHODS This cross-sectional study included 79 premenopausal women, divided in three groups of women with SSD treated with prolactin-sparing antipsychotics (n = 21) or prolactin-raising antipsychotics (n = 27), and age-matched women without SSD (n = 31). Circulating 17β-estradiol was compared among groups. In patients, we assessed the relationship between prolactin and 17β-estradiol, and the relationships of these hormones to symptom severity and cognition, using correlation analyses and backward regression models. RESULTS In women receiving prolactin-raising antipsychotics, 17β-estradiol levels were lower as compared to both other groups (H(2) = 8.34; p = 0.015), and prolactin was inversely correlated with 17β-estradiol (r=-0.42, p = 0.030). In the prolactin-raising group, 17β-estradiol correlated positively with verbal fluency (r = 0.52, p = 0.009), and 17β-estradiol and prolactin together explained 29% of the variation in processing speed (β17β-estradiol = 0.24, βprolactin = -0.45, F(2,25) = 5.98, p = 0.008). In the prolactin-sparing group, 17β-estradiol correlated negatively with depression/anxiety (r = -0.57, p = 0.014), and together with prolactin explained 26% of the variation in total symptoms (β17β-estradiol = -0.41, βprolactin = 0.32, F(2,18) = 4.44, p = 0.027). CONCLUSIONS In women with SSD, antipsychotic-induced prolactin elevation was related to lower estrogen levels. Further, estrogens negatively correlated with symptom severity and positively with cognition, whereas prolactin levels correlated negatively with cognition. Our findings stress the clinical importance of maintaining healthy levels of prolactin and estrogens in women with SSD.
Collapse
Affiliation(s)
- Bodyl A Brand
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK.
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands.
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - Janna N de Boer
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- Center for Young Children, Karakter Child and Adolescent Psychiatry, Nijmegen, The Netherlands
| | - Elske J M Willemse
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Cynthia S Weickert
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Iris E Sommer
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
- Department of Biomedical Sciences and Systems, Cognitive Neurosciences, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Thomas W Weickert
- Neuroscience Research Australia, Sydney, NSW, Australia
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
6
|
Wengler K, Trujillo P, Cassidy CM, Horga G. Neuromelanin-sensitive MRI for mechanistic research and biomarker development in psychiatry. Neuropsychopharmacology 2024; 50:137-152. [PMID: 39160355 PMCID: PMC11526017 DOI: 10.1038/s41386-024-01934-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024]
Abstract
Neuromelanin-sensitive MRI is a burgeoning non-invasive neuroimaging method with an increasing number of applications in psychiatric research. This MRI modality is sensitive to the concentration of neuromelanin, which is synthesized from intracellular catecholamines and accumulates in catecholaminergic nuclei including the dopaminergic substantia nigra and the noradrenergic locus coeruleus. Emerging data suggest the utility of neuromelanin-sensitive MRI as a proxy measure for variability in catecholamine metabolism and function, even in the absence of catecholaminergic cell loss. Given the importance of catecholamine function to several psychiatric disorders and their treatments, neuromelanin-sensitive MRI is ideally positioned as an informative and easy-to-acquire catecholaminergic index. In this review paper, we examine basic aspects of neuromelanin and neuromelanin-sensitive MRI and focus on its psychiatric applications in the contexts of mechanistic research and biomarker development. We discuss ongoing debates and state-of-the-art research into the mechanisms of the neuromelanin-sensitive MRI contrast, standardized protocols and optimized analytic approaches, and application of cutting-edge methods such as machine learning and artificial intelligence to enhance the feasibility and predictive power of neuromelanin-sensitive-MRI-based tools. We finally lay out important future directions to allow neuromelanin-sensitive-MRI to fulfill its potential as a key component of the research, and ultimately clinical, toolbox in psychiatry.
Collapse
Affiliation(s)
- Kenneth Wengler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Vanderbilt, TN, USA
| | - Clifford M Cassidy
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Guillermo Horga
- New York State Psychiatric Institute, New York, NY, USA.
- Department of Psychiatry, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Acero-Castillo MC, Correia MBM, Caixeta FV, Motta V, Barros M, Maior RS. Is the antidepressant effect of ketamine separate from its psychotomimetic effect? A review of rodent models. Neuropharmacology 2024; 258:110088. [PMID: 39032814 DOI: 10.1016/j.neuropharm.2024.110088] [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: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Ketamine is an NMDA (N-methyl-d-aspartate) glutamate receptor antagonist, which has a myriad of dose-dependent pharmacological and behavioral effects, including anesthetic, sedative, amnestic, analgesic, and anti-inflammatory properties. Intriguingly, ketamine at subanesthetic doses displays a relevant profile both in mimicking symptoms of schizophrenia and also as the first fast-acting treatment for depression. Here, we present an overview of the state-of-the-art knowledge about ketamine as an antidepressant as well as a pharmacological model of schizophrenia in animal models and human participants. Ketamine's dual effect appears to arise from its mechanism of action involving NMDA receptors, with both immediate and downstream consequences being triggered as a result. Finally, we discuss the feasibility of a unified approach linking the glutamatergic hypothesis of schizophrenia to the promising preclinical and clinical success of ketamine in the treatment of refractory depression.
Collapse
Affiliation(s)
- M C Acero-Castillo
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M B M Correia
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil; Department of Anthropology, Emory University, Atlanta GA, ZIP 30322, USA
| | - F V Caixeta
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - V Motta
- Department of Basic Psychological Processes, Institute of Psychology, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M Barros
- Department of Pharmacy, School of Health Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - R S Maior
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil.
| |
Collapse
|
8
|
Remali J, Aizat WM. Medicinal plants and plant-based traditional medicine: Alternative treatments for depression and their potential mechanisms of action. Heliyon 2024; 10:e38986. [PMID: 39640650 PMCID: PMC11620067 DOI: 10.1016/j.heliyon.2024.e38986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
Background Clinical depression is a serious public health issue that affects 4.7 % of the world's population and can lead to suicide tendencies. Although drug medications are available, only 60 % of the depressed patients respond positively to the treatments, while the rest experience side effects that resulted in the discontinuation of their medication. Thus, there is an urgent need for developing a new anti-depressant with a distinct mode of action and manageable side effects. One of the options is using medicinal plants or plant-based traditional medicine as alternative therapies for psychiatric disorders. Objectives Therefore, the objective of this review was twofold; to identify and critically evaluate anti-depressant properties of medicinal plants or those incorporated in traditional medicine; and to discuss their possible mechanism of action as well as challenges and way forward for this alternative treatment approach. Methods Relevant research articles were retrieved from various databases, including Scopus, PubMed, and Web of Science, for the period from 2018 to 2020, and the search was updated in September 2024. The inclusion criterion was relevance to antidepressants, while the exclusion criteria included duplicates, lack of full-text availability, and non-English publications. Results Through an extensive literature review, more than 40 medicinal plant species with antidepressant effects were identified, some of which are part of traditional medicine. The list of the said plant species included Albizia zygia (DC.) J.F.Macbr., Calculus bovis Sativus, Celastrus paniculatus Willd., Cinnamomum sp., Erythrina velutina Willd., Ficus platyphylla Delile, Garcinia mangostana Linn., Hyptis martiusii Benth, and Polygonum multiflorum Thunb. Anti-depressant mechanisms associated with those plants were further characterised based on their modes of action such as anti-oxidation system, anti-inflammation action, modulation of various neurotransmitters, neuroprotective effect, the regulation of hypothalamic-pituitary-adrenal (HPA) axis and anti-depressant mechanism. The challenges and future outlook of this alternative and complementary medicine are also explored and discussed. Conclusion This pool of identified plant species is hoped to offer health care professionals the best possible alternatives of anti-depressants from natural phytocompounds that are efficacious, safe and affordable for applications in future clinical settings.
Collapse
Affiliation(s)
- Juwairiah Remali
- Department of Pathology, Hospital Pulau Pinang, Jalan Residensi, 10450, George Town, Pulau Pinang, Malaysia
| | - Wan Mohd Aizat
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia (UKM), 43600, Bangi, Selangor, Malaysia
| |
Collapse
|
9
|
Vano LJ, McCutcheon RA, Rutigliano G, Kaar SJ, Finelli V, Nordio G, Wellby G, Sedlacik J, Statton B, Rabiner EA, Ye R, Veronese M, Hopkins SC, Koblan KS, Everall IP, Howes OD. Mesostriatal Dopaminergic Circuit Dysfunction in Schizophrenia: A Multimodal Neuromelanin-Sensitive Magnetic Resonance Imaging and [ 18F]-DOPA Positron Emission Tomography Study. Biol Psychiatry 2024; 96:674-683. [PMID: 38942349 DOI: 10.1016/j.biopsych.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Striatal hyperdopaminergia is implicated in the pathoetiology of schizophrenia, but how this relates to dopaminergic midbrain activity is unclear. Neuromelanin (NM)-sensitive magnetic resonance imaging provides a marker of long-term dopamine function. We examined whether midbrain NM-sensitive magnetic resonance imaging contrast-to-noise ratio (NM-CNR) was higher in people with schizophrenia than in healthy control (HC) participants and whether this correlated with dopamine synthesis capacity. METHODS One hundred fifty-four participants (schizophrenia group: n = 74, HC group: n = 80) underwent NM-sensitive magnetic resonance imaging of the substantia nigra and ventral tegmental area (SN-VTA). A subset of the schizophrenia group (n = 38) also received [18F]-DOPA positron emission tomography to measure dopamine synthesis capacity (Kicer) in the SN-VTA and striatum. RESULTS SN-VTA NM-CNR was significantly higher in patients with schizophrenia than in HC participants (effect size = 0.38, p = .019). This effect was greatest for voxels in the medial and ventral SN-VTA. In patients, SN-VTA Kicer positively correlated with SN-VTA NM-CNR (r = 0.44, p = .005) and striatal Kicer (r = 0.71, p < .001). Voxelwise analysis demonstrated that SN-VTA NM-CNR was positively associated with striatal Kicer (r = 0.53, p = .005) and that this relationship seemed strongest between the ventral SN-VTA and associative striatum in schizophrenia. CONCLUSIONS Our results suggest that NM levels are higher in patients with schizophrenia than in HC individuals, particularly in midbrain regions that project to parts of the striatum that receive innervation from the limbic and association cortices. The direct relationship between measures of NM and dopamine synthesis suggests that these aspects of schizophrenia pathophysiology are linked. Our findings highlight specific mesostriatal circuits as the loci of dopamine dysfunction in schizophrenia and thus as potential therapeutic targets.
Collapse
Affiliation(s)
- Luke J Vano
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, London, United Kingdom.
| | - Robert A McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, United Kingdom
| | - Grazia Rutigliano
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Stephen J Kaar
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Division of Psychology and Mental Health, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Valeria Finelli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Giovanna Nordio
- Department of Neuroimaging, King's College London, London, United Kingdom
| | - George Wellby
- Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jan Sedlacik
- Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Mansfield Centre for Innovation - MR Facility, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom
| | - Ben Statton
- Psychiatric Imaging Group, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Mansfield Centre for Innovation - MR Facility, MRC Laboratory of Medical Sciences, Hammersmith Hospital, London, United Kingdom
| | - Eugenii A Rabiner
- Invicro, London, United Kingdom; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Rong Ye
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, United Kingdom; The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Mattia Veronese
- Department of Neuroimaging, King's College London, London, United Kingdom; Department of Information Engineering, University of Padua, Padova, Italy
| | - Seth C Hopkins
- Sumitomo Pharma America, Inc., Marlborough, Massachusetts
| | | | - Ian P Everall
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
10
|
Wijdicks EFM, Ropper AH. Neuroleptic Malignant Syndrome. N Engl J Med 2024; 391:1130-1138. [PMID: 39321364 DOI: 10.1056/nejmra2404606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Affiliation(s)
- Eelco F M Wijdicks
- From the Neurosciences Intensive Care Unit, Mayo Clinic Hospital, Rochester, MN (E.F.M.W.)
| | - Allan H Ropper
- From the Neurosciences Intensive Care Unit, Mayo Clinic Hospital, Rochester, MN (E.F.M.W.)
| |
Collapse
|
11
|
Olivetti PR, Torres-Herraez A, Gallo ME, Raudales R, Sumerau M, Moyles S, Balsam PD, Kellendonk C. Inhibition of striatal indirect pathway during second postnatal week leads to long-lasting deficits in motivated behavior. Neuropsychopharmacology 2024:10.1038/s41386-024-01997-x. [PMID: 39327472 DOI: 10.1038/s41386-024-01997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Schizophrenia is a neuropsychiatric disorder with postulated neurodevelopmental etiology. Genetic and imaging studies have shown enhanced dopamine and D2 receptor occupancy in the striatum of patients with schizophrenia. However, whether alterations in postnatal striatal dopamine can lead to long-lasting changes in brain function and behavior is still unclear. Here, we approximated striatal D2R hyperfunction in mice via designer receptor-mediated activation of inhibitory Gi-protein signaling during a defined postnatal time window. We found that Gi-mediated inhibition of the indirect pathway (IP) during postnatal days 8-15 led to long-lasting decreases in locomotor activity and motivated behavior measured in the adult animal. In vivo photometry further showed that the motivational deficit was associated with an attenuated adaptation of outcome-evoked dopamine levels to changes in effort requirements. These data establish a sensitive time window of D2R-regulated striatal development with long-lasting impacts on neuronal function and behavior.
Collapse
Affiliation(s)
- Pedro R Olivetti
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Arturo Torres-Herraez
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Meghan E Gallo
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Ricardo Raudales
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - MaryElena Sumerau
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Barnard College Undergraduate Program, Barnard College 3009 Broadway, New York, NY, USA
| | - Sinead Moyles
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Barnard College Undergraduate Program, Barnard College 3009 Broadway, New York, NY, USA
| | - Peter D Balsam
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neuroscience and Behavior, Barnard College 3009 Broadway, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
- Department of Molecular Pharmacology & Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
12
|
Liu L, Li Z, Kong D, Huang Y, Wu D, Zhao H, Gao X, Zhang X, Yang M. Neuroimaging markers of aberrant brain activity and treatment response in schizophrenia patients based on brain complexity. Transl Psychiatry 2024; 14:365. [PMID: 39251595 PMCID: PMC11384759 DOI: 10.1038/s41398-024-03067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024] Open
Abstract
The complexity of brain activity reflects its ability to process information, adapt to environmental changes, and transition between states. However, it remains unclear how schizophrenia (SZ) affects brain activity complexity, particularly its dynamic changes. This study aimed to investigate the abnormal patterns of brain activity complexity in SZ, their relationship with cognitive deficits, and the impact of antipsychotic medication. Forty-four drug-naive first-episode (DNFE) SZ patients and thirty demographically matched healthy controls (HC) were included. Functional MRI-based sliding window analysis was utilized for the first time to calculate weighted permutation entropy to characterize complex patterns of brain activity in SZ patients before and after 12 weeks of risperidone treatment. Results revealed reduced complexity in the caudate, putamen, and pallidum at baseline in SZ patients compared to HC, with reduced complexity in the left caudate positively correlated with Continuous Performance Test (CPT) and Category Fluency Test scores. After treatment, the complexity of the left caudate increased. Regions with abnormal complexity showed decreased functional connectivity, with complexity positively correlated with connectivity strength. We observed that the dynamic complexity of the brain exhibited the characteristic of spontaneous, recurring "complexity drop", potentially reflecting transient state transitions in the resting brain. Compared to HC, patients exhibited reduced scope, intensity, and duration of complexity drop, all of which improved after treatment. Reduced duration was negatively correlated with CPT scores and positively with clinical symptoms. The results suggest that abnormalities in brain activity complexity and its dynamic changes may underlie cognitive deficits and clinical symptoms in SZ patients. Antipsychotic treatment partially restores these abnormalities, highlighting their potential as indicators of treatment efficacy and biomarkers for personalized therapy.
Collapse
Affiliation(s)
- Liju Liu
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Zezhi Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Di Kong
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yanqing Huang
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Diwei Wu
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Huachang Zhao
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xin Gao
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiangyang Zhang
- Affiliated Mental Health Center of Anhui Medical University; Hefei Fourth People's Hospital; Anhui Mental Health Center, Hefei, PR China.
| | - Mi Yang
- The Fourth People's Hospital of Chengdu, The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
13
|
Sullivan PF, Yao S, Hjerling-Leffler J. Schizophrenia genomics: genetic complexity and functional insights. Nat Rev Neurosci 2024; 25:611-624. [PMID: 39030273 DOI: 10.1038/s41583-024-00837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/21/2024]
Abstract
Determining the causes of schizophrenia has been a notoriously intractable problem, resistant to a multitude of investigative approaches over centuries. In recent decades, genomic studies have delivered hundreds of robust findings that implicate nearly 300 common genetic variants (via genome-wide association studies) and more than 20 rare variants (via whole-exome sequencing and copy number variant studies) as risk factors for schizophrenia. In parallel, functional genomic and neurobiological studies have provided exceptionally detailed information about the cellular composition of the brain and its interconnections in neurotypical individuals and, increasingly, in those with schizophrenia. Taken together, these results suggest unexpected complexity in the mechanisms that drive schizophrenia, pointing to the involvement of ensembles of genes (polygenicity) rather than single-gene causation. In this Review, we describe what we now know about the genetics of schizophrenia and consider the neurobiological implications of this information.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Aparna TK, D ST, Dar MA, Gul R, Sivasubramanian R. Polydopamine functionalized FeTiO 3 nanohexagons for selective and simultaneous electrochemical determination of dopamine and uric acid. RSC Adv 2024; 14:26694-26702. [PMID: 39184000 PMCID: PMC11340443 DOI: 10.1039/d4ra04148h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Herein we report the simultaneous detection of dopamine (DA) and uric acid (UA) using polydopamine (PDA) functionalized FeTiO3 nanohexagons. The nanohexagons were hydrothermally synthesized and subsequently functionalized with PDA in a Tris-buffer solution. The PDA functionalized nanostructure was characterized using X-ray diffraction (XRD), scanning electron microscopy (SEM), transmission electron microscopy (TEM), X-ray photoelectron spectroscopy (XPS), and Fourier transform infrared (FTIR), respectively. The SEM and TEM investigations revealed the presence of FeTiO3 nanohexagons along with a peripheral coating of PDA over the nanostructures. The XRD pattern confirmed the formation of the ilmenite structure, while the chemical structure was investigated through XPS and FTIR respectively. Using cyclic voltammetry (CV) the efficacy of FeTiO3-PDA electrode was evaluated toward DA oxidation. The enhanced activity of the functionalized electrode in DA oxidation, as compared to the untreated FeTiO3, may be attributed to the significant presence of hydroxyl, amine, and imine functional groups over the polymer layer. Differential pulse voltammetry (DPV) was utilized for the detection of DA and UA. With a linear range of 50 μM to 250 μM, the detection limits of 0.30 μM and 4.61 μM were determined for DA and UA, respectively. The peak separation of 263 mV between DA and UA demonstrates the sensor's remarkable selectivity. In addition, the study displayed the ability to detect both DA and UA simultaneously, and the validity of the sensor was evaluated in serum samples, respectively.
Collapse
Affiliation(s)
- T K Aparna
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras Chennai Tamilnadu 600036 India
| | - Swathi Tharani D
- Electrochemical Sensors and Energy Materials Lab, PSG Institute of Advanced Studies Coimbatore Tamil Nadu 641004 India
| | - Mushtaq Ahmad Dar
- Center of Excellence for Research in Engineering Materials, King Saud University Riyadh 11421 Saudi Arabia
| | - Rukhsana Gul
- Obesity Research Center, King Saud University Riyadh 11461 Saudi Arabia
| | - R Sivasubramanian
- Department of Chemistry, School of Physical Sciences, Amrita Vishwa Vidyapeetham Amaravati Andhra Pradesh 522503 India
| |
Collapse
|
15
|
Keller GB, Sterzer P. Predictive Processing: A Circuit Approach to Psychosis. Annu Rev Neurosci 2024; 47:85-101. [PMID: 38424472 DOI: 10.1146/annurev-neuro-100223-121214] [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] [Indexed: 03/02/2024]
Abstract
Predictive processing is a computational framework that aims to explain how the brain processes sensory information by making predictions about the environment and minimizing prediction errors. It can also be used to explain some of the key symptoms of psychotic disorders such as schizophrenia. In recent years, substantial advances have been made in our understanding of the neuronal circuitry that underlies predictive processing in cortex. In this review, we summarize these findings and how they might relate to psychosis and to observed cell type-specific effects of antipsychotic drugs. We argue that quantifying the effects of antipsychotic drugs on specific neuronal circuit elements is a promising approach to understanding not only the mechanism of action of antipsychotic drugs but also psychosis. Finally, we outline some of the key experiments that should be done. The aims of this review are to provide an overview of the current circuit-based approaches to psychosis and to encourage further research in this direction.
Collapse
Affiliation(s)
- Georg B Keller
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
- Faculty of Natural Science, University of Basel, Basel, Switzerland
| | - Philipp Sterzer
- Department of Psychiatry, University of Basel, Basel, Switzerland
| |
Collapse
|
16
|
Kolomeets NS, Uranova NA. Deficit of satellite oligodendrocytes of neurons in the rostral part of the head of the caudate nucleus in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01869-x. [PMID: 39073446 DOI: 10.1007/s00406-024-01869-x] [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: 03/26/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Increasing evidence implicates compromised myelin integrity and oligodendrocyte abnormalities in the dysfunction of neuronal networks in schizophrenia. We previously reported a deficiency of myelinating oligodendrocytes (OL), oligodendrocyte progenitors (OP) and satellite oligodendrocytes of neurons (Sat-OL) in the prefrontal cortex and the inferior parietal cortex - cortical hubs of the frontoparietal cognitive network and default mode network (DMN) altered in schizophrenia. Deficiency of OL and OP was also detected in the head of the caudate nucleus (HCN), which accumulates cortical projections from the associative cortex and is the central node of these networks. However, the number of Sat-Ol per neuron in schizophrenia has not been studied in the HCN. In the current study we estimated the number of Sat-Ol per neuron in the rostral part of the HCN in schizophrenia (n = 18) compared to healthy controls (n = 18) in the same section collection that was previously used to study the number Ol and OP. We found a significant decrease of the number of Sat-Ol per neuron (- 50%, p < 0.001) in schizophrenia as compared to normal controls. Considering that the rostral part of the HCN is an individual network-specific projection zone of the DMN, the deficit of Sat-Ol found in schizophrenia may be related to the dysfunctional DMN-HCN connections, which has been repeatedly described in schizophrenia. The dramatic decrease of the number of Sat-Ol per neuron may be partially related to a pronounced excess of dopamine concentration in the rostral part of the HCN in schizophrenia.
Collapse
Affiliation(s)
- N S Kolomeets
- Laboratory of Clinical Neuropathology, Mental Health Research Center, Kashirskoe shosse 34, Moscow, 115522, Russia
| | - N A Uranova
- Laboratory of Clinical Neuropathology, Mental Health Research Center, Kashirskoe shosse 34, Moscow, 115522, Russia.
| |
Collapse
|
17
|
Petersen KA, Zong W, Depoy LM, Scott MR, Shankar VG, Burns JN, Cerwensky AJ, Kim SM, Ketchesin KD, Tseng GC, McClung CA. Comparative rhythmic transcriptome profiling of human and mouse striatal subregions. Neuropsychopharmacology 2024; 49:796-805. [PMID: 38182777 PMCID: PMC10948754 DOI: 10.1038/s41386-023-01788-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024]
Abstract
The human striatum can be subdivided into the caudate, putamen, and nucleus accumbens (NAc). In mice, this roughly corresponds to the dorsal medial striatum (DMS), dorsal lateral striatum (DLS), and ventral striatum (NAc). Each of these structures have some overlapping and distinct functions related to motor control, cognitive processing, motivation, and reward. Previously, we used a "time-of-death" approach to identify diurnal rhythms in RNA transcripts in these three human striatal subregions. Here, we identify molecular rhythms across similar striatal subregions collected from C57BL/6J mice across 6 times of day and compare results to the human striatum. Pathway analysis indicates a large degree of overlap between species in rhythmic transcripts involved in processes like cellular stress, energy metabolism, and translation. Notably, a striking finding in humans is that small nucleolar RNAs (snoRNAs) and long non-coding RNAs (lncRNAs) are among the most highly rhythmic transcripts in the NAc and this is not conserved in mice, suggesting the rhythmicity of RNA processing in this region could be uniquely human. Furthermore, the peak timing of overlapping rhythmic genes is altered between species, but not consistently in one direction. Taken together, these studies reveal conserved as well as distinct transcriptome rhythms across the human and mouse striatum and are an important step in understanding the normal function of diurnal rhythms in humans and model organisms in these regions and how disruption could lead to pathology.
Collapse
Affiliation(s)
- Kaitlyn A Petersen
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lauren M Depoy
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline R Scott
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vaishnavi G Shankar
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer N Burns
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison J Cerwensky
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sam-Moon Kim
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kyle D Ketchesin
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Colleen A McClung
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Stowell R, Wang KH. Dopaminergic signaling regulates microglial surveillance and adolescent plasticity in the frontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584167. [PMID: 38559264 PMCID: PMC10979918 DOI: 10.1101/2024.03.08.584167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Adolescence is a sensitive period for frontal cortical development and cognitive maturation. The dopaminergic (DA) mesofrontal circuit is particularly malleable in response to changes in adolescent experience and DA activity. However, the cellular mechanisms engaged in this plasticity remain unexplored. Here, we report that microglia, the innate immune cells of the brain, are uniquely sensitive to adolescent mesofrontal DA signaling. Longitudinal in vivo two-photon imaging in mice shows that frontal cortical microglia respond dynamically to plasticity-inducing behavioral or optogenetic DA axon stimulation with increased parenchymal and DA bouton surveillance. Microglial-axon contact precedes new bouton formation, and both D1 and D2-type DA receptors regulate microglial-bouton interactions and axonal plasticity. Moreover, D2 antagonism in adults reinstates adolescent plasticity, including increased microglial surveillance and new DA bouton formation. Our results reveal that DA signaling regulates microglial surveillance and axonal plasticity uniquely in the adolescent frontal cortex, presenting potential interventions for restoring plasticity in the adult brain.
Collapse
Affiliation(s)
- Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| | - Kuan Hong Wang
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| |
Collapse
|
19
|
Hamati R, Ahrens J, Shvetz C, Holahan MR, Tuominen L. 65 years of research on dopamine's role in classical fear conditioning and extinction: A systematic review. Eur J Neurosci 2024; 59:1099-1140. [PMID: 37848184 DOI: 10.1111/ejn.16157] [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: 02/14/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023]
Abstract
Dopamine, a catecholamine neurotransmitter, has historically been associated with the encoding of reward, whereas its role in aversion has received less attention. Here, we systematically gathered the vast evidence of the role of dopamine in the simplest forms of aversive learning: classical fear conditioning and extinction. In the past, crude methods were used to augment or inhibit dopamine to study its relationship with fear conditioning and extinction. More advanced techniques such as conditional genetic, chemogenic and optogenetic approaches now provide causal evidence for dopamine's role in these learning processes. Dopamine neurons encode conditioned stimuli during fear conditioning and extinction and convey the signal via activation of D1-4 receptor sites particularly in the amygdala, prefrontal cortex and striatum. The coordinated activation of dopamine receptors allows for the continuous formation, consolidation, retrieval and updating of fear and extinction memory in a dynamic and reciprocal manner. Based on the reviewed literature, we conclude that dopamine is crucial for the encoding of classical fear conditioning and extinction and contributes in a way that is comparable to its role in encoding reward.
Collapse
Affiliation(s)
- Rami Hamati
- Neuroscience Graduate Program, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
| | - Jessica Ahrens
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Cecelia Shvetz
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Matthew R Holahan
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Lauri Tuominen
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Wengler K, Baker SC, Velikovskaya A, Fogelson A, Girgis RR, Reyes-Madrigal F, Lee S, de la Fuente-Sandoval C, Ojeil N, Horga G. Generalizability and Out-of-Sample Predictive Ability of Associations Between Neuromelanin-Sensitive Magnetic Resonance Imaging and Psychosis in Antipsychotic-Free Individuals. JAMA Psychiatry 2024; 81:198-208. [PMID: 37938847 PMCID: PMC10633403 DOI: 10.1001/jamapsychiatry.2023.4305] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/08/2023] [Indexed: 11/10/2023]
Abstract
Importance The link between psychosis and dopaminergic dysfunction is established, but no generalizable biomarkers with clear potential for clinical adoption exist. Objective To replicate previous findings relating neuromelanin-sensitive magnetic resonance imaging (NM-MRI), a proxy measure of dopamine function, to psychosis severity in antipsychotic-free individuals in the psychosis spectrum and to evaluate the out-of-sample predictive ability of NM-MRI for psychosis severity. Design, Setting, and Participants This cross-sectional study recruited participants from 2019 to 2023 in the New York City area (main samples) and Mexico City area (external validation sample). The main samples consisted of 42 antipsychotic-free patients with schizophrenia, 53 antipsychotic-free individuals at clinical high risk for psychosis (CHR), and 52 matched healthy controls. An external validation sample consisted of 16 antipsychotic-naive patients with schizophrenia. Main Outcomes and Measures NM-MRI contrast within a subregion of the substantia nigra previously linked to psychosis severity (a priori psychosis region of interest [ROI]) and psychosis severity measured using the Positive and Negative Syndrome Scale (PANSS) in schizophrenia and the Structured Interview for Psychosis-Risk Syndromes (SIPS) in CHR. The cross-validated performance of linear support vector regression to predict psychosis severity across schizophrenia and CHR was assessed, and a final trained model was tested on the external validation sample. Results Of the 163 included participants, 76 (46.6%) were female, and the mean (SD) age was 29.2 (10.4) years. In the schizophrenia sample, higher PANSS positive total scores correlated with higher mean NM-MRI contrast in the psychosis ROI (t37 = 2.24, P = .03; partial r = 0.35; 95% CI, 0.05 to 0.55). In the CHR sample, no significant association was found between higher SIPS positive total score and NM-MRI contrast in the psychosis ROI (t48 = -0.55, P = .68; partial r = -0.08; 95% CI, -0.36 to 0.23). The 10-fold cross-validated prediction accuracy of psychosis severity was above chance in held-out test data (mean r = 0.305, P = .01; mean root-mean-square error [RMSE] = 1.001, P = .005). External validation prediction accuracy was also above chance (r = 0.422, P = .046; RMSE = 0.882, P = .047). Conclusions and Relevance This study provided a direct ROI-based replication of the in-sample association between NM-MRI contrast and psychosis severity in antipsychotic-free patients with schizophrenia. In turn, it failed to replicate such association in CHR individuals. Most critically, cross-validated machine-learning analyses provided a proof-of-concept demonstration that NM-MRI patterns can be used to predict psychosis severity in new data, suggesting potential for developing clinically useful tools.
Collapse
Affiliation(s)
- Kenneth Wengler
- Department of Psychiatry, Columbia University, New York, New York
- New York State Psychiatric Institute, New York
| | - Seth C. Baker
- New York State Psychiatric Institute, New York
- University at Buffalo Jacobs School of Medicine and Biological Sciences, Buffalo, New York
| | | | | | - Ragy R. Girgis
- Department of Psychiatry, Columbia University, New York, New York
- New York State Psychiatric Institute, New York
| | - Francisco Reyes-Madrigal
- Laboratory of Experimental Psychiatry & Neuropsychiatry Department, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Seonjoo Lee
- Department of Psychiatry, Columbia University, New York, New York
- New York State Psychiatric Institute, New York
- Department of Biostatistics, Columbia University, New York, New York
| | - Camilo de la Fuente-Sandoval
- Laboratory of Experimental Psychiatry & Neuropsychiatry Department, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | | | - Guillermo Horga
- Department of Psychiatry, Columbia University, New York, New York
- New York State Psychiatric Institute, New York
| |
Collapse
|
21
|
Guiard BP, Gotti G. The High-Precision Liquid Chromatography with Electrochemical Detection (HPLC-ECD) for Monoamines Neurotransmitters and Their Metabolites: A Review. Molecules 2024; 29:496. [PMID: 38276574 PMCID: PMC10818480 DOI: 10.3390/molecules29020496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
This review highlights the advantages of high-precision liquid chromatography with an electrochemical detector (HPLC-ECD) in detecting and quantifying biological samples obtained through intracerebral microdialysis, specifically the serotonergic and dopaminergic systems: Serotonin (5-HT), 5-hydroxyindolacetic acid (5-HIAA), 3,4-dihydroxyphenylacetic acid (DOPAC), dopamine (DA), 3-metoxytryptamin (3-MT) and homovanillic acid (HVA). Recognized for its speed and selectivity, HPLC enables direct analysis of intracerebral microdialysis samples without complex derivatization. Various chromatographic methods, including reverse phase (RP), are explored for neurotransmitters (NTs) and metabolites separation. Electrochemical detector (ECD), particularly with glassy carbon (GC) electrodes, is emphasized for its simplicity and sensitivity, aimed at enhancing reproducibility through optimization strategies such as modified electrode materials. This paper underscores the determination of limits of detection (LOD) and quantification (LOQ) and the linear range (L.R.) showcasing the potential for real-time monitoring of compounds concentrations. A non-exhaustive compilation of literature values for LOD, LOQ, and L.R. from recent publications is included.
Collapse
Affiliation(s)
- Bruno P. Guiard
- Centre de Recherches sur la Cognition Animale (CRCA), CNRS UMR5169, 31062 Toulouse, France;
- Centre de Biologie Intégrative (CBI), Faculté Sciences Ingénierie (FSI), Université de Toulouse III, 31062 Toulouse, France
| | - Guillaume Gotti
- Centre de Recherches sur la Cognition Animale (CRCA), CNRS UMR5169, 31062 Toulouse, France;
- Centre de Biologie Intégrative (CBI), Faculté Sciences Ingénierie (FSI), Université de Toulouse III, 31062 Toulouse, France
| |
Collapse
|
22
|
Kristensen TD, Raghava JM, Skjerbæk MW, Dhollander T, Syeda W, Ambrosen KS, Bojesen KB, Nielsen MØ, Pantelis C, Glenthøj BY, Ebdrup BH. Fibre density and fibre-bundle cross-section of the corticospinal tract are distinctly linked to psychosis-specific symptoms in antipsychotic-naïve patients with first-episode schizophrenia. Eur Arch Psychiatry Clin Neurosci 2023; 273:1797-1812. [PMID: 37012463 PMCID: PMC10713712 DOI: 10.1007/s00406-023-01598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023]
Abstract
Multiple lines of research support the dysconnectivity hypothesis of schizophrenia. However, findings on white matter (WM) alterations in patients with schizophrenia are widespread and non-specific. Confounding factors from magnetic resonance image (MRI) processing, clinical diversity, antipsychotic exposure, and substance use may underlie some of the variability. By application of refined methodology and careful sampling, we rectified common confounders investigating WM and symptom correlates in a sample of strictly antipsychotic-naïve first-episode patients with schizophrenia. Eighty-six patients and 112 matched controls underwent diffusion MRI. Using fixel-based analysis (FBA), we extracted fibre-specific measures such as fibre density and fibre-bundle cross-section. Group differences on fixel-wise measures were examined with multivariate general linear modelling. Psychopathology was assessed with the Positive and Negative Syndrome Scale. We separately tested multivariate correlations between fixel-wise measures and predefined psychosis-specific versus anxio-depressive symptoms. Results were corrected for multiple comparisons. Patients displayed reduced fibre density in the body of corpus callosum and in the middle cerebellar peduncle. Fibre density and fibre-bundle cross-section of the corticospinal tract were positively correlated with suspiciousness/persecution, and negatively correlated with delusions. Fibre-bundle cross-section of isthmus of corpus callosum and hallucinatory behaviour were negatively correlated. Fibre density and fibre-bundle cross-section of genu and splenium of corpus callosum were negative correlated with anxio-depressive symptoms. FBA revealed fibre-specific properties of WM abnormalities in patients and differentiated associations between WM and psychosis-specific versus anxio-depressive symptoms. Our findings encourage an itemised approach to investigate the relationship between WM microstructure and clinical symptoms in patients with schizophrenia.
Collapse
Affiliation(s)
- Tina D Kristensen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark.
| | - Jayachandra M Raghava
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Glostrup, Denmark
| | - Martin W Skjerbæk
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
| | - Thijs Dhollander
- Developmental Imaging, Murdoch Children's Research Institute, Victoria, Australia
| | - Warda Syeda
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne and Melbourne Health, Victoria, Australia
| | - Karen S Ambrosen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
| | - Kirsten B Bojesen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
| | - Mette Ø Nielsen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christos Pantelis
- Developmental Imaging, Murdoch Children's Research Institute, Victoria, Australia
| | - Birte Y Glenthøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bjørn H Ebdrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Centre Glostrup, Copenhagen University Hospital, Nordstjernevej 41, 2600, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Blaess S, Krabbe S. Cell type specificity for circuit output in the midbrain dopaminergic system. Curr Opin Neurobiol 2023; 83:102811. [PMID: 37972537 DOI: 10.1016/j.conb.2023.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Midbrain dopaminergic neurons are a relatively small group of neurons in the mammalian brain controlling a wide range of behaviors. In recent years, increasingly sophisticated tracing, imaging, transcriptomic, and machine learning approaches have provided substantial insights into the anatomical, molecular, and functional heterogeneity of dopaminergic neurons. Despite this wealth of new knowledge, it remains unclear whether and how the diverse features defining dopaminergic subclasses converge to delineate functional ensembles within the dopaminergic system. Here, we review recent studies investigating various aspects of dopaminergic heterogeneity and discuss how development, behavior, and disease influence subtype characteristics. We then outline what further approaches could be pursued to gain a more inclusive picture of dopaminergic diversity, which could be crucial to understanding the functional architecture of this system.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Sabine Krabbe
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
24
|
Mastwal S, Li X, Stowell R, Manion M, Zhang W, Kim NS, Yoon KJ, Song H, Ming GL, Wang KH. Adolescent neurostimulation of dopamine circuit reverses genetic deficits in frontal cortex function. eLife 2023; 12:RP87414. [PMID: 37830916 PMCID: PMC10575630 DOI: 10.7554/elife.87414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Dopamine system dysfunction is implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive and novel paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in Arc or Disc1 to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc-/- mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in Disc1+/- mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Xinjian Li
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Matthew Manion
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Nam-Shik Kim
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Ki-Jun Yoon
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Guo-Li Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| |
Collapse
|
25
|
Chen Y, Wang X, Xiao B, Luo Z, Long H. Mechanisms and Functions of Activity-Regulated Cytoskeleton-Associated Protein in Synaptic Plasticity. Mol Neurobiol 2023; 60:5738-5754. [PMID: 37338805 DOI: 10.1007/s12035-023-03442-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/10/2023] [Indexed: 06/21/2023]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is one of the most important regulators of cognitive functions in the brain regions. As a hub protein, Arc plays different roles in modulating synaptic plasticity. Arc supports the maintenance of long-term potentiation (LTP) by regulating actin cytoskeletal dynamics, while it guides the endocytosis of AMPAR in long-term depression (LTD). Moreover, Arc can self-assemble into capsids, leading to a new way of communicating among neurons. The transcription and translation of the immediate early gene Arc are rigorous procedures guided by numerous factors, and RNA polymerase II (Pol II) is considered to regulate the precise timing dynamics of gene expression. Since astrocytes can secrete brain-derived neurotrophic factor (BDNF) and L-lactate, their unique roles in Arc expression are emphasized. Here, we review the entire process of Arc expression and summarize the factors that can affect Arc expression and function, including noncoding RNAs, transcription factors, and posttranscriptional regulations. We also attempt to review the functional states and mechanisms of Arc in modulating synaptic plasticity. Furthermore, we discuss the recent progress in understanding the roles of Arc in the occurrence of major neurological disorders and provide new thoughts for future research on Arc.
Collapse
Affiliation(s)
- Yifan Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Xiangya School of Stomatology, Central South University, Changsha, 410008, Hunan, China
| | - Xiaohu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, Hunan, People's Republic of China, 410008.
| | - Hongyu Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, Hunan, People's Republic of China, 410008.
| |
Collapse
|
26
|
Managò F, Scheggia D, Pontillo M, Mereu M, Mastrogiacomo R, Udayan G, Valentini P, Tata MC, Weinberger DR, Weickert CS, Pompa PP, De Luca MA, Vicari S, Papaleo F. Dopaminergic signalling and behavioural alterations by Comt-Dtnbp1 genetic interaction and their clinical relevance. Br J Pharmacol 2023; 180:2514-2531. [PMID: 37218669 DOI: 10.1111/bph.16147] [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: 06/13/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Cognitive and motor functions are modulated by dopaminergic signalling, which is shaped by several genetic factors. The biological effects of single genetic variants might differ depending on epistatic interactions that can be functionally multi-directional and non-linear. EXPERIMENTAL APPROACH We performed behavioural and neurochemical assessments in genetically modified mice and behavioural assessments and genetic screening in human patients with 22q11.2 deletion syndrome (22q11.2DS). KEY RESULTS Here, we confirm a genetic interaction between the Comt (catechol-O-methyltransferase, human orthologue: COMT) and Dtnbp1 (dystrobrevin binding protein 1, alias dysbindin, human orthologue: DTNBP1) genes that modulate cortical and striatal dopaminergic signalling in a manner not predictable by the effects of each single gene. In mice, Comt-by-Dtnbp1 concomitant reduction leads to a hypoactive mesocortical and a hyperactive mesostriatal dopamine pathway, associated with specific cognitive abnormalities. Like mice, in subjects with the 22q11.2DS (characterized by COMT hemideletion and dopamine alterations), COMT-by-DTNBP1 concomitant reduction was associated with analogous cognitive disturbances. We then developed an easy and inexpensive colourimetric kit for the genetic screening of common COMT and DTNBP1 functional genetic variants for clinical application. CONCLUSIONS AND IMPLICATIONS These findings illustrate an epistatic interaction of two dopamine-related genes and their functional effects, supporting the need to address genetic interaction mechanisms at the base of complex behavioural traits.
Collapse
Affiliation(s)
- Francesca Managò
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Diego Scheggia
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Pontillo
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maddalena Mereu
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rosa Mastrogiacomo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Gayatri Udayan
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Paola Valentini
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Cynthia S Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Pier Paolo Pompa
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Maria A De Luca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Vicari
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
27
|
Gao N, Liu Z, Wang H, Shen C, Dong Z, Cui W, Xiong WC, Mei L. Deficiency of Cullin 3, a Protein Encoded by a Schizophrenia and Autism Risk Gene, Impairs Behaviors by Enhancing the Excitability of Ventral Tegmental Area (VTA) DA Neurons. J Neurosci 2023; 43:6249-6267. [PMID: 37558490 PMCID: PMC10490515 DOI: 10.1523/jneurosci.0247-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/09/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
The dopaminergic neuromodulator system is fundamental to brain functions. Abnormal dopamine (DA) pathway is implicated in psychiatric disorders, including schizophrenia (SZ) and autism spectrum disorder (ASD). Mutations in Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex, have been associated with SZ and ASD. However, little is known about the function and mechanism of CUL3 in the DA system. Here, we show that CUL3 is critical for the function of DA neurons and DA-relevant behaviors in male mice. CUL3-deficient mice exhibited hyperactive locomotion, deficits in working memory and sensorimotor gating, and increased sensitivity to psychostimulants. In addition, enhanced DA signaling and elevated excitability of the VTA DA neurons were observed in CUL3-deficient animals. Behavioral impairments were attenuated by dopamine D2 receptor antagonist haloperidol and chemogenetic inhibition of DA neurons. Furthermore, we identified HCN2, a hyperpolarization-activated and cyclic nucleotide-gated channel, as a potential target of CUL3 in DA neurons. Our study indicates that CUL3 controls DA neuronal activity by maintaining ion channel homeostasis and provides insight into the role of CUL3 in the pathogenesis of psychiatric disorders.SIGNIFICANCE STATEMENT This study provides evidence that Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex that has been associated with autism spectrum disorder and schizophrenia, controls the excitability of dopamine (DA) neurons in mice. Its DA-specific heterozygous deficiency increased spontaneous locomotion, impaired working memory and sensorimotor gating, and elevated response to psychostimulants. We showed that CUL3 deficiency increased the excitability of VTA DA neurons, and inhibiting D2 receptor or DA neuronal activity attenuated behavioral deficits of CUL3-deficient mice. We found HCN2, a hyperpolarization-activated channel, as a target of CUL3 in DA neurons. Our findings reveal CUL3's role in DA neurons and offer insights into the pathogenic mechanisms of autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Zhipeng Liu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Chen Shen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106
- Chinese Institutes for Medical Research, Beijing, China 100069
- Capital Medical University, Beijing, China 100069
| |
Collapse
|
28
|
Wakabayashi C, Kunugi H. Possible involvement of Interleukin-17A in the deterioration of prepulse inhibition on acoustic startle response in mice. Neuropsychopharmacol Rep 2023; 43:365-372. [PMID: 37280178 PMCID: PMC10496063 DOI: 10.1002/npr2.12351] [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: 01/05/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/08/2023] Open
Abstract
AIM Proinflammatory cytokines such as interleukin-6 (IL-6) and IL-17A have been implicated in the pathophysiology of schizophrenia which often shows sensorimotor gating abnormalities. This study aimed to examine whether a proinflammatory cytokine, IL-17A, induces impairment in sensorimotor gating in mice. We also examined whether IL-17A administration affects GSK3α/β protein level or phosphorylation in the striatum. METHODS Recombinant mouse IL-17A (low-dose: 0.5 ng/mL and high-dose: 50 ng/mL with 10 μL/g mouse body weight, respectively) or vehicle was intraperitoneally administered into C57BL/6 male mice 10 times in 3 weeks (sub-chronic administration). Prepulse inhibition test using acoustic startle stimulus was conducted 4 weeks after the final IL-17A administration. We evaluated the effect of IL-17A administration on protein level or phosphorylation of GSK3α/β in the striatum by using Western blot analysis. RESULTS Administration of IL-17A induced significant PPI deterioration. Low-dose of IL-17A administration significantly decreased both GSK3α (Ser21) and GSK3β (Ser9) phosphorylation in mouse striatum. There was no significant alteration of GSK3α/β protein levels except for GSK3α in low-dose IL-17A administration group. CONCLUSION We demonstrated for the first time that sub-chronic IL-17A administration induced PPI disruption and that IL-17A administration resulted in decreased phosphorylation of GSKα/β at the striatum. These results suggest that IL-17A could be a target molecule in the prevention and treatment of sensorimotor gating abnormalities observed in schizophrenia.
Collapse
Affiliation(s)
- Chisato Wakabayashi
- Department of Mental Disorder Research, National Institute of NeuroscienceNational Center of Neurology and PsychiatryKodairaJapan
- Faculty of Pharmaceutical SciencesHimeji Dokkyo UniversityHimejiJapan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of NeuroscienceNational Center of Neurology and PsychiatryKodairaJapan
- Department of PsychiatryTeikyo University School of MedicineItabashiJapan
| |
Collapse
|
29
|
García-Yagüe ÁJ, Cuadrado A. Mechanisms of NURR1 Regulation: Consequences for Its Biological Activity and Involvement in Pathology. Int J Mol Sci 2023; 24:12280. [PMID: 37569656 PMCID: PMC10419244 DOI: 10.3390/ijms241512280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
NURR1 (Nuclear receptor-related 1 protein or NR4A2) is a nuclear protein receptor transcription factor with an essential role in the development, regulation, and maintenance of dopaminergic neurons and mediates the response to stressful stimuli during the perinatal period in mammalian brain development. The dysregulation of NURR1 activity may play a role in various diseases, including the onset and progression of neurodegenerative diseases, and several other pathologies. NURR1 is regulated by multiple mechanisms, among which phosphorylation by kinases or SUMOylation are the best characterized. Both post-translational modifications can regulate the activity of NURR1, affecting its stability and transcriptional activity. Other non-post-translational regulatory mechanisms include changes in its subcellular distribution or interaction with other protein partners by heterodimerization, also affecting its transcription activity. Here, we summarize the currently known regulatory mechanisms of NURR1 and provide a brief overview of its participation in pathological alterations.
Collapse
Affiliation(s)
- Ángel Juan García-Yagüe
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28027 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-CIBERNED), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta, 28029 Madrid, Spain
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28027 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-CIBERNED), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta, 28029 Madrid, Spain
| |
Collapse
|
30
|
Jayanti S, Dalla Verde C, Tiribelli C, Gazzin S. Inflammation, Dopaminergic Brain and Bilirubin. Int J Mol Sci 2023; 24:11478. [PMID: 37511235 PMCID: PMC10380707 DOI: 10.3390/ijms241411478] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Dopamine is a well-known neurotransmitter due to its involvement in Parkinson's disease (PD). Dopamine is not only involved in PD but also controls multiple mental and physical activities, such as the pleasure of food, friends and loved ones, music, art, mood, cognition, motivation, fear, affective disorders, addiction, attention deficit disorder, depression, and schizophrenia. Dopaminergic neurons (DOPAn) are susceptible to stressors, and inflammation is a recognized risk for neuronal malfunctioning and cell death in major neurodegenerative diseases. Less is known for non-neurodegenerative conditions. Among the endogenous defenses, bilirubin, a heme metabolite, has been shown to possess important anti-inflammatory activity and, most importantly, to prevent DOPAn demise in an ex vivo model of PD by acting on the tumor necrosis factor-alpha (TNFα). This review summarizes the evidence linking DOPAn, inflammation (when possible, specifically TNFα), and bilirubin as an anti-inflammatory in order to understand what is known, the gaps that need filling, and the hypotheses of anti-inflammatory strategies to preserve dopamine homeostasis with bilirubin included.
Collapse
Affiliation(s)
- Sri Jayanti
- Italian Liver Foundation, Liver Brain Unit "Rita Moretti", Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Camilla Dalla Verde
- Italian Liver Foundation, Liver Brain Unit "Rita Moretti", Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy
| | - Claudio Tiribelli
- Italian Liver Foundation, Liver Brain Unit "Rita Moretti", Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy
| | - Silvia Gazzin
- Italian Liver Foundation, Liver Brain Unit "Rita Moretti", Area Science Park, Bldg. Q, SS 14, Km 163,5, 34149 Trieste, Italy
| |
Collapse
|
31
|
Mastwal S, Li X, Stowell R, Manion M, Zhang W, Kim NS, Yoon KJ, Song H, Ming GL, Wang KH. Adolescent neurostimulation of dopamine circuit reverses genetic deficits in frontal cortex function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.526987. [PMID: 36778456 PMCID: PMC9915739 DOI: 10.1101/2023.02.03.526987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dopamine system dysfunction is commonly implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive to such pharmacological treatments and novel research paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in the Arc or DISC1 genes to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc mutant mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in DISC1 mutant mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, Bethesda, MD 20892
| | - Xinjian Li
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, Bethesda, MD 20892
| | - Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| | - Matthew Manion
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, Bethesda, MD 20892
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, Bethesda, MD 20892
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| | - Nam-Shik Kim
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ki-jun Yoon
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Guo-li Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, Bethesda, MD 20892
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
32
|
Weerasinghe DK, Hodge JM, Pasco JA, Samarasinghe RM, Azimi Manavi B, Williams LJ. Antipsychotic-induced bone loss: the role of dopamine, serotonin and adrenergic receptor signalling. Front Cell Dev Biol 2023; 11:1184550. [PMID: 37305679 PMCID: PMC10248006 DOI: 10.3389/fcell.2023.1184550] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Antipsychotics are commonly used in treating psychiatric disorders. These medications primarily target dopamine the serotonin receptors, they have some affinity to adrenergic, histamine, glutamate and muscarinic receptors. There is clinical evidence that antipsychotic use decreases BMD and increases fracture risk, with dopamine, serotonin and adrenergic receptor-signalling becoming an increasing area of focus where the presence of these receptors in osteoclasts and osteoblasts have been demonstrated. Osteoclasts and osteoblasts are the most important cells in the bone remodelling and the bone regeneration process where the activity of these cells determine the bone resorption and formation process in order to maintain healthy bone. However, an imbalance in osteoclast and osteoblast activity can lead to decreased BMD and increased fracture risk, which is also believed to be exacerbated by antipsychotics use. Therefore, the aim of this review is to provide an overview of the mechanisms of action of first, second and third generation antipsychotics and the expression profiles of dopamine, serotonin and adrenergic receptors during osteoclastogenesis and osteoblastogenesis.
Collapse
Affiliation(s)
- D. Kavindi Weerasinghe
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Jason M. Hodge
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| | - Julie A. Pasco
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
- Department of Medicine—Western Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Rasika M. Samarasinghe
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Behnaz Azimi Manavi
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Lana J. Williams
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
- Barwon Health, Geelong, VIC, Australia
| |
Collapse
|
33
|
Lotan A, Luza S, Opazo CM, Ayton S, Lane DJR, Mancuso S, Pereira A, Sundram S, Weickert CS, Bousman C, Pantelis C, Everall IP, Bush AI. Perturbed iron biology in the prefrontal cortex of people with schizophrenia. Mol Psychiatry 2023; 28:2058-2070. [PMID: 36750734 PMCID: PMC10575779 DOI: 10.1038/s41380-023-01979-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Despite loss of grey matter volume and emergence of distinct cognitive deficits in young adults diagnosed with schizophrenia, current treatments for schizophrenia do not target disruptions in late maturational reshaping of the prefrontal cortex. Iron, the most abundant transition metal in the brain, is essential to brain development and function, but in excess, it can impair major neurotransmission systems and lead to lipid peroxidation, neuroinflammation and accelerated aging. However, analysis of cortical iron biology in schizophrenia has not been reported in modern literature. Using a combination of inductively coupled plasma-mass spectrometry and western blots, we quantified iron and its major-storage protein, ferritin, in post-mortem prefrontal cortex specimens obtained from three independent, well-characterised brain tissue resources. Compared to matched controls (n = 85), among schizophrenia cases (n = 86) we found elevated tissue iron, unlikely to be confounded by demographic and lifestyle variables, by duration, dose and type of antipsychotic medications used or by copper and zinc levels. We further observed a loss of physiologic age-dependent iron accumulation among people with schizophrenia, in that the iron level among cases was already high in young adulthood. Ferritin, which stores iron in a redox-inactive form, was paradoxically decreased in individuals with the disorder. Such iron-ferritin uncoupling could alter free, chemically reactive, tissue iron in key reasoning and planning areas of the young-adult schizophrenia cortex. Using a prediction model based on iron and ferritin, our data provide a pathophysiologic link between perturbed cortical iron biology and schizophrenia and indicate that achievement of optimal cortical iron homeostasis could offer a new therapeutic target.
Collapse
Affiliation(s)
- Amit Lotan
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Psychiatry and the Biological Psychiatry Laboratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Serafino Mancuso
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Avril Pereira
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Mental Health Program, Monash Health, Melbourne, VIC, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
| | - Ian P Everall
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
34
|
Tabata H, Mori D, Matsuki T, Yoshizaki K, Asai M, Nakayama A, Ozaki N, Nagata KI. Histological Analysis of a Mouse Model of the 22q11.2 Microdeletion Syndrome. Biomolecules 2023; 13:biom13050763. [PMID: 37238632 DOI: 10.3390/biom13050763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
22q11.2 deletion syndrome (22q11.2DS) is associated with a high risk of developing various psychiatric and developmental disorders, including schizophrenia and early-onset Parkinson's disease. Recently, a mouse model of this disease, Del(3.0Mb)/+, mimicking the 3.0 Mb deletion which is most frequently found in patients with 22q11.2DS, was generated. The behavior of this mouse model was extensively studied and several abnormalities related to the symptoms of 22q11.2DS were found. However, the histological features of their brains have been little addressed. Here we describe the cytoarchitectures of the brains of Del(3.0Mb)/+ mice. First, we investigated the overall histology of the embryonic and adult cerebral cortices, but they were indistinguishable from the wild type. However, the morphologies of individual neurons were slightly but significantly changed from the wild type counterparts in a region-specific manner. The dendritic branches and/or dendritic spine densities of neurons in the medial prefrontal cortex, nucleus accumbens, and primary somatosensory cortex were reduced. We also observed reduced axon innervation of dopaminergic neurons into the prefrontal cortex. Given these affected neurons function together as the dopamine system to control animal behaviors, the impairment we observed may explain a part of the abnormal behaviors of Del(3.0Mb)/+ mice and the psychiatric symptoms of 22q11.2DS.
Collapse
Affiliation(s)
- Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya 466-8550, Japan
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Kaichi Yoshizaki
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Masato Asai
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Atsuo Nakayama
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa-ku, Nagoya 464-0814, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
35
|
De Simone G, Mazza B, Vellucci L, Barone A, Ciccarelli M, de Bartolomeis A. Schizophrenia Synaptic Pathology and Antipsychotic Treatment in the Framework of Oxidative and Mitochondrial Dysfunction: Translational Highlights for the Clinics and Treatment. Antioxidants (Basel) 2023; 12:antiox12040975. [PMID: 37107350 PMCID: PMC10135787 DOI: 10.3390/antiox12040975] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Schizophrenia is a worldwide mental illness characterized by alterations at dopaminergic and glutamatergic synapses resulting in global dysconnectivity within and between brain networks. Impairments in inflammatory processes, mitochondrial functions, energy expenditure, and oxidative stress have been extensively associated with schizophrenia pathophysiology. Antipsychotics, the mainstay of schizophrenia pharmacological treatment and all sharing the common feature of dopamine D2 receptor occupancy, may affect antioxidant pathways as well as mitochondrial protein levels and gene expression. Here, we systematically reviewed the available evidence on antioxidants' mechanisms in antipsychotic action and the impact of first- and second-generation compounds on mitochondrial functions and oxidative stress. We further focused on clinical trials addressing the efficacy and tolerability of antioxidants as an augmentation strategy of antipsychotic treatment. EMBASE, Scopus, and Medline/PubMed databases were interrogated. The selection process was conducted in respect of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria. Several mitochondrial proteins involved in cell viability, energy metabolism, and regulation of oxidative systems were reported to be significantly modified by antipsychotic treatment with differences between first- and second-generation drugs. Finally, antioxidants may affect cognitive and psychotic symptoms in patients with schizophrenia, and although the evidence is only preliminary, the results indicate that further studies are warranted.
Collapse
Affiliation(s)
- Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
- UNESCO Chair on Health Education and Sustainable Development, University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
36
|
Oldehinkel M, Tiego J, Sabaroedin K, Chopra S, Francey SM, O'Donoghue B, Cropley V, Nelson B, Graham J, Baldwin L, Yuen HP, Allott K, Alvarez-Jimenez M, Harrigan S, Pantelis C, Wood SJ, McGorry P, Bellgrove MA, Fornito A. Gradients of striatal function in antipsychotic-free first-episode psychosis and schizotypy. Transl Psychiatry 2023; 13:128. [PMID: 37072388 PMCID: PMC10113219 DOI: 10.1038/s41398-023-02417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/20/2023] Open
Abstract
Both psychotic illness and subclinical psychosis-like experiences (PLEs) have been associated with cortico-striatal dysfunction. This work has largely relied on a discrete parcellation of the striatum into distinct functional areas, but recent evidence suggests that the striatum comprises multiple overlapping and smoothly varying gradients (i.e., modes) of functional organization. Here, we investigated two of these functional connectivity modes, previously associated with variations in the topographic patterning of cortico-striatal connectivity (first-order gradient), and dopaminergic innervation of the striatum (second-order gradient), and assessed continuities in striatal function from subclinical to clinical domains. We applied connectopic mapping to resting-state fMRI data to obtain the first-order and second-order striatal connectivity modes in two distinct samples: (1) 56 antipsychotic-free patients (26 females) with first-episode psychosis (FEP) and 27 healthy controls (17 females); and (2) a community-based cohort of 377 healthy individuals (213 females) comprehensively assessed for subclinical PLEs and schizotypy. The first-order "cortico-striatal" and second-order "dopaminergic" connectivity gradients were significantly different in FEP patients compared to controls bilaterally. In the independent sample of healthy individuals, variations in the left first-order "cortico-striatal" connectivity gradient were associated with inter-individual differences in a factor capturing general schizotypy and PLE severity. The presumed cortico-striatal connectivity gradient was implicated in both subclinical and clinical cohorts, suggesting that variations in its organization may represent a neurobiological trait marker across the psychosis continuum. Disruption of the presumed dopaminergic gradient was only noticeable in patients, suggesting that neurotransmitter dysfunction may be more apparent to clinical illness.
Collapse
Affiliation(s)
- Marianne Oldehinkel
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands.
| | - Jeggan Tiego
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Kristina Sabaroedin
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Sidhant Chopra
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Shona M Francey
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | | | - Vanessa Cropley
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Barnaby Nelson
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | | | - Lara Baldwin
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | | | - Kelly Allott
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Mario Alvarez-Jimenez
- Orygen Youth Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
| | - Susy Harrigan
- Department of Social Work, Monash University, Melbourne, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Stephen J Wood
- Orygen Youth Health, Parkville, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, Australia
- School of Psychology, University of Birmingham, Birmingham, UK
| | - Patrick McGorry
- Orygen Youth Health, Parkville, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Mark A Bellgrove
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Clayton, Australia
| |
Collapse
|
37
|
Brown NK, Roche JK, Farmer CB, Roberts RC. Evidence for upregulation of excitatory synaptic transmission in the substantia nigra in Schizophrenia: a postmortem ultrastructural study. J Neural Transm (Vienna) 2023; 130:561-573. [PMID: 36735096 DOI: 10.1007/s00702-023-02593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/14/2023] [Indexed: 02/04/2023]
Abstract
The dopamine hypothesis of schizophrenia suggests that psychotic symptoms originate from dysregulation of dopaminergic activity, which may be controlled by upstream innervation. We hypothesized that we would find anatomical evidence for the hyperexcitability seen in the SN. We examined and quantified synaptic morphology, which correlates with function, in the postmortem substantia nigra (SN) from 15 schizophrenia and 12 normal subjects. Synapses were counted using stereological techniques and classified based on the morphology of the post-synaptic density (PSD) and the presence or absence of a presynaptic density. The density and proportion of excitatory synapses was higher in the schizophrenia group than in controls, while the proportion (but not density) of inhibitory synapses was lower. We also detected in the schizophrenia group an increase in density of synapses with a PSD of intermediate thickness, which may represent excitatory synapses. The density of synapses with presynaptic densities was similar in both groups. The density of synapses with mixed morphologies was higher in the schizophrenia group than in controls. The human SN contains atypical synaptic morphology. We found an excess amount and proportion of excitatory synapses in the SN in schizophrenia that could result in hyperactivity and drive the psychotic symptoms of schizophrenia. The sources of afferent excitatory inputs to the SN arise from the subthalamic nucleus, the pedunculopontine nucleus, and the ventral tegmental area (VTA), areas that could be the source of excess excitation. Synapses with mixed morphologies may represent inputs from the VTA, which release multiple transmitters.
Collapse
Affiliation(s)
- Nicole K Brown
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Joy K Roche
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Charlene B Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA
| | - Rosalinda C Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 835C, 1720 2nd Avenue South, Birmingham, AL, 35294, USA.
| |
Collapse
|
38
|
Hudon A, Couture J, Dellazizzo L, Beaudoin M, Phraxayavong K, Potvin S, Dumais A. Dyadic Interactions of Treatment-Resistant Schizophrenia Patients Having Followed Virtual Reality Therapy: A Content Analysis. J Clin Med 2023; 12:jcm12062299. [PMID: 36983300 PMCID: PMC10053204 DOI: 10.3390/jcm12062299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023] Open
Abstract
(1) Background: Very little is known about the inner therapeutic processes of psychotherapy interventions for patients suffering from treatment-resistant schizophrenia. Avatar therapy (AT) is one such modalities in which the patient is undergoing immersive sessions in which they interact with an Avatar representing their main persistent auditory verbal hallucination. The aim of this study is to identify the most prevalent dyadic interactions between the patient and the Avatar in AT for patient’s suffering from TRS. (2) Methods: A content analysis of 256 verbatims originating from 32 patients who completed AT between 2017 and 2022 at the Institut universitaire en santé mentale de Montréal was conducted to identify dyadic interactions between the patients and their Avatar. (3) Results: Five key dyads were identified to occur on average more than 10 times for each participant during the immersive sessions across their AT: (Avatar: Reinforcement, Patient: Self-affirmation), (Avatar: Provocation, Patient: Self-affirmation), (Avatar: Coping mechanisms, Patient: Prevention), (Patient: Self-affirmation, Avatar: Reinforcement), and (Patient: Self-appraisal, Avatar: Reinforcement). (4) Conclusion: These dyads offer a first qualitative insight to the interpersonal dynamics and patient-avatar relationships taking place during AT. Future studies on the implication of such dyadic interactions with the therapeutic outcome of AT should be conducted considering the importance of dyadic relationships in psychotherapy.
Collapse
Affiliation(s)
- Alexandre Hudon
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montréal, QC H1N 3J4, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Jonathan Couture
- Faculty of Medicine, Department of Medicine, Campus Montréal, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Laura Dellazizzo
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montréal, QC H1N 3J4, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mélissa Beaudoin
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montréal, QC H1N 3J4, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | - Stéphane Potvin
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montréal, QC H1N 3J4, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Alexandre Dumais
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Montréal, Montréal, QC H1N 3J4, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Services et Recherches Psychiatriques AD, Montréal, QC H1C 1H1, Canada
- Institut National de Psychiatrie Légale Philippe-Pinel, Montréal, QC H1C 1H1, Canada
- Correspondence:
| |
Collapse
|
39
|
Tomita Y, Suzuki K, Yamasaki S, Toriumi K, Miyashita M, Ando S, Endo K, Yoshikawa A, Tabata K, Usami S, Hiraiwa-Hasegawa M, Itokawa M, Kawaji H, Kasai K, Nishida A, Arai M. Urinary exosomal microRNAs as predictive biomarkers for persistent psychotic-like experiences. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:14. [PMID: 36906656 PMCID: PMC10008540 DOI: 10.1038/s41537-023-00340-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
Psychotic-like experiences (PLEs) occur occasionally in adolescence and mostly disappear with increasing age. Their presence, if persistent, is considered a robust risk factor for subsequent psychiatric disorders. To date, only a few biological markers have been investigated for persistent PLE prediction. This study identified urinary exosomal microRNAs that can serve as predictive biomarkers for persistent PLEs. This study was part of a population-based biomarker subsample study of the Tokyo Teen Cohort Study. A total of 345 participants aged 13 (baseline) and 14 (follow-up) years underwent PLE assessments by experienced psychiatrists using semi-structured interviews. We defined remitted and persistent PLEs based on longitudinal profiles. We obtained urine at baseline and the expression levels of urinary exosomal miRNAs were compared between 15 individuals with persistent PLEs and 15 age- and sex-matched individuals with remitted PLEs. We constructed a logistic regression model to examine whether miRNA expression levels could predict persistent PLEs. We identified six significant differentially expressed microRNAs, namely hsa-miR-486-5p, hsa-miR-199a-3p, hsa-miR-144-5p, hsa-miR-451a, hsa-miR-143-3p, and hsa-miR-142-3p. The predictive model showed an area under the curve of 0.860 (95% confidence interval: 0.713-0.993) for five-fold cross-validation. We found a subset of urinary exosomal microRNAs that were differentially expressed in persistent PLEs and presented the likelihood that a microRNA-based statistical model could predict them with high accuracy. Therefore, urine exosomal miRNAs may serve as novel biomarkers for the risk of psychiatric disorders.
Collapse
Affiliation(s)
- Yasufumi Tomita
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Suzuki
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry, Shinshu University School of Medicine, Matsumoto, Japan.,Department of Community Mental Health, Shinshu University School of Medicine, Matsumoto, Japan
| | - Syudo Yamasaki
- Unit for Mental Health Promotion, Research Center for Social Science and Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuya Toriumi
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mitsuhiro Miyashita
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Unit for Mental Health Promotion, Research Center for Social Science and Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shuntaro Ando
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaori Endo
- Unit for Mental Health Promotion, Research Center for Social Science and Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akane Yoshikawa
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichi Tabata
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Usami
- Center for Research and Development on Transition from Secondary to Higher Education, The University of Tokyo, Tokyo, Japan
| | - Mariko Hiraiwa-Hasegawa
- Department of Evolutionary Studies of Biosystems, The Graduate University for the Advanced Studies, SOKENDAI, Hayama, Japan
| | - Masanari Itokawa
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Hideya Kawaji
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, University of Tokyo, Tokyo, Japan
| | - Atsushi Nishida
- Unit for Mental Health Promotion, Research Center for Social Science and Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Makoto Arai
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
40
|
Treatment-Resistant Schizophrenia, Clozapine Resistance, Genetic Associations, and Implications for Precision Psychiatry: A Scoping Review. Genes (Basel) 2023; 14:genes14030689. [PMID: 36980961 PMCID: PMC10048540 DOI: 10.3390/genes14030689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Treatment-resistant schizophrenia (TRS) is often associated with severe burden of disease, poor quality of life and functional impairment. Clozapine is the gold standard for the treatment of TRS, although it is also known to cause significant side effects in some patients. In view of the burgeoning interest in the role of genetic factors in precision psychiatry, we conducted a scoping review to narratively summarize the current genetic factors associated with TRS, clozapine resistance and side effects to clozapine treatment. We searched PubMed from inception to December 2022 and included 104 relevant studies in this review. Extant evidence comprised associations between TRS and clozapine resistance with genetic factors related to mainly dopaminergic and serotoninergic neurotransmitter systems, specifically, TRS and rs4680, rs4818 within COMT, and rs1799978 within DRD2; clozapine resistance and DRD3 polymorphisms, CYP1A2 polymorphisms; weight gain with LEP and SNAP-25 genes; and agranulocytosis risk with HLA-related polymorphisms. Future studies, including replication in larger multi-site samples, are still needed to elucidate putative risk genes and the interactions between different genes and their correlations with relevant clinical factors such as psychopathology, psychosocial functioning, cognition and progressive changes with treatment over time in TRS and clozapine resistance.
Collapse
|
41
|
Dang X, Liu J, Zhang Z, Luo XJ. Mendelian Randomization Study Using Dopaminergic Neuron-Specific eQTL Identifies Novel Risk Genes for Schizophrenia. Mol Neurobiol 2023; 60:1537-1546. [PMID: 36517655 DOI: 10.1007/s12035-022-03160-3] [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: 06/06/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022]
Abstract
Multiple integrative studies have been performed to identify the potential target genes of the non-coding schizophrenia (SCZ) risk variants. However, all the integrative studies used expression quantitative trait loci (eQTL) data from bulk tissues. Considering the cell type-specific regulatory effect of many genetic variants, it is important to conduct integrative studies using cell type-specific eQTL data. Here, we conduct a Mendelian randomization (MR) study by integrating genome-wide associations of SCZ (74,776 cases and 101,023 controls) and eQTL data (N = 215) from dopaminergic neurons, which were differentiated from human-induced pluripotent stem cell (iPSC) lines. For eQTL from young post-mitotic dopaminergic neurons (differentiation of iPSC for 30 days, D30), we identified 34 genes whose genetically regulated expression in dopaminergic neurons may have a causal role in SCZ. Among which, ARL3 showed the most significant associations with SCZ. For eQTL from more mature dopaminergic neurons (D52), we identified 37 potential SCZ causal genes, and ARL3 and GNL3 showed the most significant associations. Only 12 genes showed significant associations with SCZ in both D30 and D52 eQTL datasets, indicating the time point-specific genetic regulatory effects in young post-mitotic dopaminergic neurons and more mature dopaminergic neurons. Comparing the results from dopaminergic neurons with bulk brain tissues prioritized 2 high-confidence risk genes, including DDHD2 and GALNT10. Our study identifies multiple risk genes whose genetically regulated expression in dopaminergic neurons may have a causal role in SCZ. Further mechanistic investigation will provide pivotal insights into SCZ pathophysiology.
Collapse
Affiliation(s)
- Xinglun Dang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Zhijun Zhang
- Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
- Department of Neurology, School of Medicine, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Xiong-Jian Luo
- Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China.
- Department of Neurology, School of Medicine, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, Southeast University, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
42
|
Brandl F, Knolle F, Avram M, Leucht C, Yakushev I, Priller J, Leucht S, Ziegler S, Wunderlich K, Sorg C. Negative symptoms, striatal dopamine and model-free reward decision-making in schizophrenia. Brain 2023; 146:767-777. [PMID: 35875972 DOI: 10.1093/brain/awac268] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/13/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Negative symptoms, such as lack of motivation or social withdrawal, are highly prevalent and debilitating in patients with schizophrenia. Underlying mechanisms of negative symptoms are incompletely understood, thereby preventing the development of targeted treatments. We hypothesized that in patients with schizophrenia during psychotic remission, impaired influences of both model-based and model-free reward predictions on decision-making ('reward prediction influence', RPI) underlie negative symptoms. We focused on psychotic remission, because psychotic symptoms might confound reward-based decision-making. Moreover, we hypothesized that impaired model-based/model-free RPIs depend on alterations of both associative striatum dopamine synthesis and storage (DSS) and executive functioning. Both factors influence RPI in healthy subjects and are typically impaired in schizophrenia. Twenty-five patients with schizophrenia with pronounced negative symptoms during psychotic remission and 24 healthy controls were included in the study. Negative symptom severity was measured by the Positive and Negative Syndrome Scale negative subscale, model-based/model-free RPI by the two-stage decision task, associative striatum DSS by 18F-DOPA positron emission tomography and executive functioning by the symbol coding task. Model-free RPI was selectively reduced in patients and associated with negative symptom severity as well as with reduced associative striatum DSS (in patients only) and executive functions (both in patients and controls). In contrast, model-based RPI was not altered in patients. Results provide evidence for impaired model-free reward prediction influence as a mechanism for negative symptoms in schizophrenia as well as for reduced associative striatum dopamine and executive dysfunction as relevant factors. Data suggest potential treatment targets for patients with schizophrenia and pronounced negative symptoms.
Collapse
Affiliation(s)
- Felix Brandl
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, 81675, Germany
| | - Franziska Knolle
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,Department of Psychiatry, University of Cambridge, Cambridge CB20SZ, UK
| | - Mihai Avram
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany
| | - Claudia Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, 81675, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, 81675, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,Neuropsychiatry, Charité-Universitätsmedizin Berlin, and DZNE, Berlin, 10117, Germany.,UK DRI at University of Edinburgh, Edinburgh EH16 4SB, UK.,IoPPN, King's College London, London SE5 8AF, UK
| | - Stefan Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,Department of Psychosis studies, King's College London, London, UK
| | - Sibylle Ziegler
- Department of Nuclear Medicine, Ludwig-Maximilians University Munich, Munich, 81377, Germany
| | - Klaus Wunderlich
- Department of Psychology, Ludwig-Maximilians University Munich, Munich, 81377, Germany
| | - Christian Sorg
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, 81675, Germany.,TUM-NIC Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, 81675, Germany
| |
Collapse
|
43
|
Fišar Z. Biological hypotheses, risk factors, and biomarkers of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110626. [PMID: 36055561 DOI: 10.1016/j.pnpbp.2022.110626] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/19/2022]
Abstract
Both the discovery of biomarkers of schizophrenia and the verification of biological hypotheses of schizophrenia are an essential part of the process of understanding the etiology of this mental disorder. Schizophrenia has long been considered a neurodevelopmental disease whose symptoms are caused by impaired synaptic signal transduction and brain neuroplasticity. Both the onset and chronic course of schizophrenia are associated with risk factors-induced disruption of brain function and the establishment of a new homeostatic setpoint characterized by biomarkers. Different risk factors and biomarkers can converge to the same symptoms of schizophrenia, suggesting that the primary cause of the disease can be highly individual. Schizophrenia-related biomarkers include measurable biochemical changes induced by stress (elevated allostatic load), mitochondrial dysfunction, neuroinflammation, oxidative and nitrosative stress, and circadian rhythm disturbances. Here is a summary of selected valid biological hypotheses of schizophrenia formulated based on risk factors and biomarkers, neurodevelopment, neuroplasticity, brain chemistry, and antipsychotic medication. The integrative neurodevelopmental-vulnerability-neurochemical model is based on current knowledge of the neurobiology of the onset and progression of the disease and the effects of antipsychotics and psychotomimetics and reflects the complex and multifactorial nature of schizophrenia.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Czech Republic.
| |
Collapse
|
44
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
45
|
Accelerated atrophy in dopaminergic targets and medial temporo-parietal regions precedes the onset of delusions in patients with Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2023; 273:229-241. [PMID: 35554669 PMCID: PMC9958148 DOI: 10.1007/s00406-022-01417-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/11/2022] [Indexed: 12/21/2022]
Abstract
People with Alzheimer's disease (AD) and delusions have worse quality of life and prognosis. However, early markers of delusions have not been identified yet. The present study investigated whether there are any detectable differences in grey matter (GM) volume and cognitive changes in the year before symptom onset between patients with AD who did and did not develop delusions. Two matched samples of AD patients, 63 who did (PT-D) and 63 who did not develop delusions (PT-ND) over 1 year, were identified from the Alzheimer's Disease Neuroimaging Initiative database. The Neuropsychiatric Inventory (NPI) was used to assess the presence of delusions. Sixty-three additional matched healthy controls (HC) were selected. Repeated-measures ANCOVA models were used to investigate group-by-time effects on the volume of selected GM regions of interest and on cognitive performance. No neurocognitive differences were observed between patient groups prior to symptom onset. Greater episodic memory decline and GM loss in bilateral caudate nuclei, medio-temporal and midline cingulo-parietal regions were found in the PT-D compared with the PT-ND group. A pattern of faster GM loss in brain areas typically affected by AD and in cortical and subcortical targets of dopaminergic pathways, paralleled by worsening of episodic memory and behavioural symptoms, may explain the emergence of delusions in patients with AD.
Collapse
|
46
|
Morris EL, Taylor SF, Kang J. On predictability of individual functional connectivity networks from clinical characteristics. Hum Brain Mapp 2022; 43:5250-5265. [PMID: 35811395 PMCID: PMC9812246 DOI: 10.1002/hbm.26000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/07/2022] [Accepted: 06/06/2022] [Indexed: 01/15/2023] Open
Abstract
In recent years, understanding functional brain connectivity has become increasingly important as a scientific tool with potential clinical implications. Statistical methods, such as graphical models and network analysis, have been adopted to construct functional connectivity networks for single subjects. Here we focus on studying the association between functional connectivity networks and clinical characteristics such as psychiatric symptoms and diagnoses. Utilizing machine learning algorithms, we propose a method to examine predictability of functional connectivity networks from clinical characteristics. Our methods can identify salient clinical characteristics predictive of the whole brain network or specific subnetworks. We illustrate our methods on the analysis of fMRI data in the Philadelphia Neurodevelopmental Cohort study, demonstrating clinically meaningful results.
Collapse
Affiliation(s)
- Emily L. Morris
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | | | - Jian Kang
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
47
|
Bio-behavioural changes in treatment-resistant socially isolated FSL rats show variable or improved response to combined fluoxetine-olanzapine versus olanzapine treatment. IBRO Neurosci Rep 2022; 13:284-298. [PMID: 36204253 PMCID: PMC9529672 DOI: 10.1016/j.ibneur.2022.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/14/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022] Open
|
48
|
Roberts RC, McCollum LA, Schoonover KE, Mabry SJ, Roche JK, Lahti AC. Ultrastructural evidence for glutamatergic dysregulation in schizophrenia. Schizophr Res 2022; 249:4-15. [PMID: 32014360 PMCID: PMC7392793 DOI: 10.1016/j.schres.2020.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/14/2022]
Abstract
The aim of this paper is to summarize ultrastructural evidence for glutamatergic dysregulation in several linked regions in postmortem schizophrenia brain. Following a brief summary of glutamate circuitry and how synapses are identified at the electron microscopic (EM) level, we will review EM pathology in the cortex and basal ganglia. We will include the effects of antipsychotic drugs and the relation of treatment response. We will discuss how these findings support or confirm other postmortem findings as well as imaging results. Briefly, synaptic and mitochondrial density in anterior cingulate cortex was decreased in schizophrenia, versus normal controls (NCs), in a selective layer specific pattern. In dorsal striatum, increases in excitatory synaptic density were detected in caudate matrix, a compartment associated with cognitive and motor function, and in the putamen patches, a region associated with limbic function and in the core of the nucleus accumbens. Patients who were treatment resistant or untreated had significantly elevated numbers of excitatory synapses in limbic striatal areas in comparison to NCs and responders. Protein levels of vGLUT2, found in subcortical glutamatergic neurons, were increased in the nucleus accumbens in schizophrenia. At the EM level, schizophrenia subjects had an increase in density of excitatory synapses in several areas of the basal ganglia. In the substantia nigra, the protein levels of vGLUT2 were elevated in untreated patients compared to NCs. The density of inhibitory synapses was decreased in schizophrenia versus NCs. In schizophrenia, glutamatergic synapses are differentially affected depending on the brain region, treatment status, and treatment response.
Collapse
Affiliation(s)
- Rosalinda C Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America.
| | - Lesley A McCollum
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Kirsten E Schoonover
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Samuel J Mabry
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Joy K Roche
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294, United States of America
| |
Collapse
|
49
|
Du X, Wei X, Ding H, Yu Y, Xie Y, Ji Y, Zhang Y, Chai C, Liang M, Li J, Zhuo C, Yu C, Qin W. Unraveling schizophrenia replicable functional connectivity disruption patterns across sites. Hum Brain Mapp 2022; 44:156-169. [PMID: 36222054 PMCID: PMC9783440 DOI: 10.1002/hbm.26108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 02/05/2023] Open
Abstract
Functional connectivity (FC) disruption is a remarkable characteristic of schizophrenia. However, heterogeneous patterns reported across sites severely hindered its clinical generalization. Based on qualified nodal-based FC of 340 schizophrenia patients (SZ) and 348 normal controls (NC) acquired from seven different scanners, this study compared four commonly used site-effect correction methods in removing the site-related heterogeneities, and then tried to cluster the abnormal FCs into several replicable and independent disrupted subnets across sites, related them to clinical symptoms, and evaluated their potentials in schizophrenia classification. Among the four site-related heterogeneity correction methods, ComBat harmonization (F1 score: 0.806 ± 0.145) achieved the overall best balance between sensitivity and false discovery rate in unraveling the aberrant FCs of schizophrenia in the local and public data sets. Hierarchical clustering analysis identified three replicable FC disruption subnets across the local and public data sets: hypo-connectivity within sensory areas (Net1), hypo-connectivity within thalamus, striatum, and ventral attention network (Net2), and hyper-connectivity between thalamus and sensory processing system (Net3). Notably, the derived composite FC within Net1 was negatively correlated with hostility and disorientation in the public validation set (p < .05). Finally, the three subnet-specific composite FCs (Best area under the receiver operating characteristic curve [AUC] = 0.728) can robustly and meaningfully discriminate the SZ from NC with comparable performance with the full identified FCs features (best AUC = 0.765) in the out-of-sample public data set (Z = -1.583, p = .114). In conclusion, ComBat harmonization was most robust in detecting aberrant connectivity for schizophrenia. Besides, the three subnet-specific composite FC measures might be replicable neuroimaging markers for schizophrenia.
Collapse
Affiliation(s)
- Xiaotong Du
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Xiaotong Wei
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Hao Ding
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina,School of Medical ImagingTianjin Medical UniversityTianjinChina
| | - Ying Yu
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Yingying Xie
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Yi Ji
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Yu Zhang
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Chao Chai
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| | - Meng Liang
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina,School of Medical ImagingTianjin Medical UniversityTianjinChina
| | - Jie Li
- Department of Psychiatry Functional Neuroimaging LaboratoryTianjin Mental Health Center, Tianjin Anding HospitalTianjinChina
| | - Chuanjun Zhuo
- Department of Psychiatry Functional Neuroimaging LaboratoryTianjin Mental Health Center, Tianjin Anding HospitalTianjinChina
| | - Chunshui Yu
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina,School of Medical ImagingTianjin Medical UniversityTianjinChina
| | - Wen Qin
- Department of RadiologyTianjin Medical University General HospitalTianjinChina,Tianjin Key Lab of Functional ImagingTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
50
|
Mastrogiacomo R, Trigilio G, Devroye C, Dautan D, Ferretti V, Losi G, Caffino L, Orso G, Marotta R, Maltese F, Vitali E, Piras G, Forgiarini A, Pacinelli G, Lia A, Rothmond DA, Waddington JL, Drago F, Fumagalli F, Luca MAD, Leggio GM, Carmignoto G, Weickert CS, Managò F, Papaleo F. Dysbindin-1A modulation of astrocytic dopamine and basal ganglia dependent behaviors relevant to schizophrenia. Mol Psychiatry 2022; 27:4201-4217. [PMID: 35821415 DOI: 10.1038/s41380-022-01683-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
The mechanisms underlying the dichotomic cortical/basal ganglia dopaminergic abnormalities in schizophrenia are unclear. Astrocytes are important non-neuronal modulators of brain circuits, but their role in dopaminergic system remains poorly explored. Microarray analyses, immunohistochemistry, and two-photon laser scanning microscopy revealed that Dys1 hypofunction increases the reactivity of astrocytes, which express only the Dys1A isoform. Notably, behavioral and electrochemical assessments in mice selectively lacking the Dys1A isoform unraveled a more prominent impact of Dys1A in behavioral and dopaminergic/D2 alterations related to basal ganglia, but not cortical functioning. Ex vivo electron microscopy and protein expression analyses indicated that selective Dys1A disruption might alter intracellular trafficking in astrocytes, but not in neurons. In agreement, Dys1A disruption only in astrocytes resulted in decreased motivation and sensorimotor gating deficits, increased astrocytic dopamine D2 receptors and decreased dopaminergic tone within basal ganglia. These processes might have clinical relevance because the caudate, but not the cortex, of patients with schizophrenia shows a reduction of the Dys1A isoform. Therefore, we started to show a hitherto unknown role for the Dys1A isoform in astrocytic-related modulation of basal ganglia behavioral and dopaminergic phenotypes, with relevance to schizophrenia.
Collapse
Affiliation(s)
- Rosa Mastrogiacomo
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Gabriella Trigilio
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Céline Devroye
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Daniel Dautan
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valentina Ferretti
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Gabriele Losi
- Neuroscience Institute, CNR, Padova, Italy.,Department of Biomedical Science, University of Padova, Padova, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Roberto Marotta
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Federica Maltese
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Enrica Vitali
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Gessica Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Alessia Forgiarini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Giada Pacinelli
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Annamaria Lia
- Neuroscience Institute, CNR, Padova, Italy.,Department of Biomedical Science, University of Padova, Padova, Italy
| | - Debora A Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | - John L Waddington
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, CNR, Padova, Italy.,Department of Biomedical Science, University of Padova, Padova, Italy
| | - Cynthia S Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | - Francesca Managò
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.
| | - Francesco Papaleo
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy. .,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
| |
Collapse
|