1
|
Gkotsoulias DG, Rullmann M, Schmitt S, Bujanow A, Zientek F, Messerschmidt K, Pampel A, Büttner AP, Schildan A, Sabri O, Müller-Vahl K, Barthel H, Möller HE. Abnormalities of iron homeostasis and the dopaminergic system in Tourette syndrome revealed by 7T MRI and PET. Brain Commun 2025; 7:fcaf104. [PMID: 40177529 PMCID: PMC11961303 DOI: 10.1093/braincomms/fcaf104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Abstract
While the implication of a dysfunctional dopaminergic system in Tourette syndrome (TS) is well established, the underlying pathophysiological mechanisms remain unclear. Apart from neurotransmitters, disturbed iron homeostasis and iron regulatory mechanisms are also suspected. Iron is a trace element of fundamental biological importance and is involved in the synthesis and metabolism of dopamine and its receptors and transporters. The goal of the current pre-registered, multi-modal, cross-sectional study was to investigate the relationship between potential iron homeostasis imbalances and dopaminergic system disturbances in patients with TS. Susceptibility-sensitive MRI at 7 Tesla was used to obtain surrogate measures for local brain iron in 25 patients with TS (age 30 ± 9 years, 6 female) and 40 matched control subjects. Additionally, dopamine D1 receptor availability was investigated with [11C]SCH23390 PET in a subgroup of 20 patients and 20 controls. Significantly reduced sub-cortical magnetic susceptibility, indicating reduced iron levels, was observed in TS patients in the caudate, pallidum, sub-thalamic nucleus, thalamus, red nucleus and substantia nigra. These reductions were accompanied by significant reductions of the [11C]SCH23390 binding potential indicating reduced availability of D1 receptors in the dorsal striatum. The D1 receptor abnormality correlated with tic severity. These results point to alterations of intra-synaptic dopamine release and reduced striatal D1 receptor binding, supporting the notion of disruption in multiple functional elements of the dopaminergic system. Such dopaminergic abnormalities appear to be associated with disturbances in iron homeostasis.
Collapse
Affiliation(s)
| | - Michael Rullmann
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig 04103, Germany
| | - Simon Schmitt
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover 30625, Germany
| | - Anna Bujanow
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
| | - Franziska Zientek
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig 04103, Germany
| | | | - André Pampel
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
| | | | - Andreas Schildan
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig 04103, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig 04103, Germany
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover 30625, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University Medical Center, Leipzig 04103, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig 04103, Germany
- Felix Bloch Institute for Solid State Physics, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
2
|
Liang B, Zhou Y, Jiang C, Zhao T, Qin D, Gao F. Role and related mechanisms of non-invasive brain stimulation in the treatment of Tourette syndrome. Brain Res Bull 2025; 222:111258. [PMID: 39954818 DOI: 10.1016/j.brainresbull.2025.111258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Tourette syndrome (TS) is a neurodevelopmental disorder characterized by impaired or delayed functional development. Although the pathology of TS remains to be determined, the continuous development of science and technology has provided new perspectives to understand its pathological mechanism. Research into non-invasive brain stimulation (NIBS) techniques, such as transcranial magnetic stimulation and direct current stimulation, have shown promising therapeutic potential in clinical studies. Furthermore, NIBS has been shown to affect the brain of patients with TS, including synaptic transmission, release of neurotransmitters, in addition to the activation of microglial cells and astrocytes. However, an exploration of the innate mechanisms is still lacking. This review aims to summarize the pathogenesis of TS and intervention with NIBS in clinical patients with TS. It aims to provide a theoretical basis for more in-depth investigations of innovative therapies for TS in the future.
Collapse
Affiliation(s)
- Boshen Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Yang Zhou
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Chengting Jiang
- Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China
| | - Ting Zhao
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Disease, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China.
| | - Fabao Gao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, China.
| |
Collapse
|
3
|
Wu X, Hao J, Jiang K, Wu M, Zhao X, Zhang X. Neuroinflammation and pathways that contribute to tourette syndrome. Ital J Pediatr 2025; 51:63. [PMID: 40022157 PMCID: PMC11871796 DOI: 10.1186/s13052-025-01874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Tourette syndrome (TS), a neurological and psychological disease, typically exhibit motor and phonic tics. The pathophysiology of TS remains controversial. Currently, the recognized pathogenesis of TS is the imbalance of neurotransmitters, involving abnormality of the cortex-striatum-thalamus-cortex circuit. Recently, clinical researches demonstrate that triggers such as infection and allergic reaction could lead to the onset or exacerbation of tic symptoms. Current studies have also suggested that neural-immune crosstalk caused by inflammation is also associated with TS, potentially leading to the occurrence of tics by inducing neurotransmitter abnormalities. Herein, we review inflammation-related factors contributing to the occurrence of TS as well as the mechanisms by which immune-inflammatory pathways mediate the onset of TS. This aims to clarify the pathogenesis of TS and provide a theoretical basis for the treatment of TS.
Collapse
Affiliation(s)
- Xinnan Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Keyu Jiang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhao
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Gunturu S, Saeidi M, Alzein O, Jafari K, Salehi M, Jaka S. Unpacking Tourette Syndrome in Children: Insights into Prevalence and Comorbidities from NSCH Data. J Clin Med 2025; 14:1485. [PMID: 40094948 PMCID: PMC11900335 DOI: 10.3390/jcm14051485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Tourette syndrome is a neurodevelopmental disorder characterized by multiple motor and vocal tics. Although Tourette syndrome is known to have various comorbidities, comprehensive data on its prevalence and associated conditions in a large, diverse population are limited. This study aimed to examine the prevalence of Tourette syndrome and its comorbidities in children aged 3 to 17 years using data from the 2021 National Survey of Children's Health (NSCH). Methods: Data from 79,236 children aged 3-17 years were analyzed. The prevalence of Tourette syndrome was assessed, and its association with socio-demographic factors and comorbid conditions, including prematurity and low birth weight, was examined using univariate and multivariate logistic regression models. Results: The prevalence of Tourette syndrome was 0.3% among children aged 3-17 years, with higher rates in males (74%) and adolescents aged 11-17 years (74%). Prematurity and low birth weight were associated with higher rates of Tourette syndrome and its comorbidities. Neurodevelopmental conditions such as ADHD (49% in Tourette syndrome vs. 10.2% in non-Tourette syndrome), autism spectrum disorder (21% vs. 3.2%), and learning disabilities were significantly more prevalent among children with Tourette syndrome. Similarly, psychiatric disorders such as anxiety (60% vs. 11.3%) and depression (25% vs. 5%) were more common in the Tourette syndrome group. Immune-based conditions, including asthma and allergies, and physical health conditions such as diabetes and vision or hearing problems, were also significantly associated with TS. Conclusions: The study highlights the significant burden of comorbidities in children with Tourette syndrome, emphasizing the need for early diagnosis and comprehensive management strategies to address the multifaceted challenges faced by these children.
Collapse
Affiliation(s)
- Sasidhar Gunturu
- Department of Psychiatry, Bronx Care Health System, Bronx, NY 10457, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahdieh Saeidi
- Department of Psychiatry, Bronx Care Health System, Bronx, NY 10457, USA
| | - Omar Alzein
- Department of Psychiatry, Bronx Care Health System, Bronx, NY 10457, USA
| | - Kamyar Jafari
- Department of Psychiatry, Bronx Care Health System, Bronx, NY 10457, USA
| | - Mona Salehi
- Department of Psychiatry, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sanobar Jaka
- Department of Psychiatry and Behavioral Sciences, Nassau University of Medical Center, East Meadow, NY 11554, USA
| |
Collapse
|
5
|
Lin L, Ruan Z, Li Y, Qiu H, Deng C, Qian L, Cui W, Tang W, Yang Z, Cheng Y, Liang Y, Su S. Brain Iron Alteration in Pediatric Tourette Syndrome: A Quantitative Susceptibility Mapping Study. Eur J Neurol 2025; 32:e70054. [PMID: 39895224 PMCID: PMC11788536 DOI: 10.1111/ene.70054] [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: 08/14/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The cortico-striato-thalamo-cortical circuits play a crucial role in the pathogenesis of Tourette syndrome (TS). While iron deficiency has been reported in adult TS, the iron content in pediatric TS remains poorly understood. This study aims to quantitatively assess whole-brain iron deposition in pediatric TS compared to typically developing (TD) children using quantitative susceptibility mapping (QSM). METHODS In this prospective study, we recruited 50 children with a clinical diagnosis of TS and 50 age- and gender-matched TD controls. Whole-brain images were acquired using 3D T1 and multi-echo gradient-recalled echo sequences. QSM maps were generated using the STISuite toolbox. After normalizing the QSM maps to Montreal Neurological Institute space, voxel-based analysis was applied to compare between-group differences in iron content. Additionally, we evaluated the relationship between iron content and tic severity in TS children using the Pearson's correlation test. RESULTS Compared to TD children, those with TS exhibited iron deficiency in the right anterior cingulum (pFDR < 0.001). Conversely, increased QSM values were observed in the bilateral putamen of TS children (pFDR < 0.001). Notably, QSM values in the left putamen showed a significant negative correlation with tic severity (p = 0.044). CONCLUSIONS Our findings suggest that disturbed brain iron homeostasis in specific regions is associated with pediatric TS. These results reinforce the importance of the cortico-striato-thalamo-cortical circuits in TS pathogenesis and highlight the potential role of iron dysregulation. Furthermore, our study demonstrates that QSM could serve as a valuable auxiliary biomarker for diagnosing and potentially monitoring pediatric TS.
Collapse
Affiliation(s)
- Liping Lin
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhibin Ruan
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yufen Li
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Huaqiong Qiu
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Chengfen Deng
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Long Qian
- Department of Biomedical Engineering, College of EngineeringPeking UniversityBeijingChina
| | - Wei Cui
- Department of Biomedical Engineering, College of EngineeringPeking UniversityBeijingChina
| | - Wen Tang
- Department of PediatricThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhiyun Yang
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yanglei Cheng
- Department of EndocrineThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yujian Liang
- Department of PediatricThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Shu Su
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
6
|
Wohlgemuth JB, Watson KH, Gill KD, Isaacs DA. Premonitory urge in tic disorders - a scoping review. Front Psychiatry 2025; 16:1504442. [PMID: 39950174 PMCID: PMC11821575 DOI: 10.3389/fpsyt.2025.1504442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Premonitory urges are uncomfortable bodily sensations preceding tics. They are highly prevalent, frequently bothersome, and increasingly recognized as a central phenotypic feature in tic disorder populations. This scoping review aimed to systematically consolidate published knowledge and identify knowledge gaps regarding premonitory urges in primary tic disorders. Methods Search strategies were deployed in five databases and five topic-relevant journals. Two independent reviewers screened all candidate abstracts against predefined inclusion criteria. One hundred and fifty-five articles were included in the scoping review. The same two reviewers independently extracted and consolidated pertinent data from included articles. Results Multiple methods for assessing premonitory urge were identified, each with strengths and weaknesses. The subjective quality of premonitory urges varies between individuals, with increased prevalence of a "not just right" urge quality in individuals with comorbid obsessive-compulsive disorder. Awareness of premonitory urge appears to arise several years after tic-onset, yet many individuals perceive their tics as voluntary responses to premonitory urges. Premonitory urges and tics are temporally coupled in real time, but premonitory urge severity and tic severity, as assessed by clinical scales, are not consistently associated. The mechanistic and developmental relationship between premonitory urges and tics remains unclear. Data are limited on premonitory urge response to treatment, but several promising interventions were identified. The insula and supplementary motor area are the neuroanatomical structures most strongly implicated in emergence of the premonitory urge. Discussion Knowledge of the clinical characteristics, measurement, and neural mechanisms of premonitory urge has advanced considerably in recent years, but important knowledge gaps remain in each of these domains. Addressing these knowledge gaps will be key to developing effective interventions for premonitory urge. Systematic Review Registration Open Science Framework (OSF) https://doi.org/10.17605/OSF.IO/WT43Z.
Collapse
Affiliation(s)
- John B. Wohlgemuth
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Kelly H. Watson
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kayce D. Gill
- Annette and Irwin Eskind Family Biomedical Library and Learning Center, Vanderbilt University, Nashville, TN, United States
| | - David A. Isaacs
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
7
|
Yang Y, Zhou J, Yang H, Wang A, Tian Y, Luo R. Structural and functional alterations in the brain gray matter among Tourette syndrome patients: a multimodal meta-analysis of fMRI and VBM studies. J Neurol 2025; 272:133. [PMID: 39812838 PMCID: PMC11735548 DOI: 10.1007/s00415-024-12852-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] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Tourette syndrome (TS) is a prevalent neurodevelopmental disorder with an uncertain etiology. Numerous neuroimaging studies have investigated patients with TS, but their conclusions remain inconsistent. The current study attempted to provide an unbiased statistical meta-analysis of published neuroimaging studies of TS. METHODS A comprehensive literature search was conducted to identify voxel-based whole-brain morphology (VBM) and functional magnetic resonance imaging (fMRI) studies related to TS. Two separate meta-analyses of neurofunctional activation and gray matter volume (GMV) were performed using a seed-point-based d-mapping software package, followed by joint and subgroup analyses. RESULTS 11 VBM studies and 18 fMRI studies were included in this study. We found that grey matter volumes were significantly decreased in the right anterior cingulate/paracingulate gyri and the left postcentral gyrus; while the cerebellum, bilateral cortico-spinal projections, and striatum showed increased GMV in patients with TS. In fMRI studies, patients with TS showed overactivation in the right superior frontal gyrus and right superior temporal gyrus, and significant hypoactivation in left SMA. In the multimodal studies, TS patients showed that there was an overlap between decreased GMV and hypoactivation in the right median cingulate/paracingulate gyri. CONCLUSION Abnormal alterations in the structure and function of the brain regions may play a role in the pathogenesis of TS in patients, and may be used as an imaging indicator for patients with TS to be diagnosed.
Collapse
Affiliation(s)
- Yue Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Jielan Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Hua Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Anqi Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Yu Tian
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China.
| | - Rong Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Frick LR. Neuroglia in Tourette syndrome and obsessive-compulsive disorder. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:325-334. [PMID: 40148053 DOI: 10.1016/b978-0-443-19102-2.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
In recent years, neuroglia have drawn the attention of researchers in the fields of neurology and psychiatry. Besides their well-known functions providing support to neurons, myelinating axons, and clearing up debris, a constantly growing of evidence indicates that glial cells are key contributors to the pathophysiology of neuropsychiatric disorders. Alterations in microglia, astrocytes, and oligodendrocytes have been described in Tourette syndrome (TS) and obsessive-compulsive disorder (OCD). The sudden onset of tics and OCD-like symptoms after infection in children (Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infections) suggests a connection with the immune system; in fact, neuroinflammation has been reported. Many imaging studies revealed abnormal myelination in the brain of TS and OCD patients, highlighting the implication of oligodendroglia in the connectivity alterations. Moreover, animal models have unveiled a cell-autonomous role of microglia and astrocytes in the etiology of pathologic grooming, which links these glial cells to the related disorder trichotillomania. This chapter reviews the state of the art and current gaps in the literature, proposing possible pathomechanisms and future research directions.
Collapse
Affiliation(s)
- Luciana R Frick
- Departments of Neurology and Medicine, Neuroscience Graduate Program, Jacobs School of Medicine & Biomedical Sciences, State University of New York at Buffalo, Clinical and Translational Research Center, Buffalo, NY, United States.
| |
Collapse
|
9
|
Szejko N, Saramak K, Müller-Vahl KR. The Use of Cannabis-Based Medicine in Selected Neurological Disorders. Curr Top Behav Neurosci 2024. [PMID: 39739176 DOI: 10.1007/7854_2024_564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Cannabis-based medicine (CBM) is used in a wide variety of different neurological disorders. While the use of CBM in the treatment of pain, AIDS wasting, loss of appetite, and spasticity is well established, CBM application in movement disorders and neurodegenerative disorders is still an emerging topic. The purpose of this chapter is to summarize current evidence behind the use of CBM in selected neurological diseases, mainly movement and neurodegenerative disorders. The best evidence for efficacy of CBM is for Tourette syndrome resulting in an improvement of tics and psychiatric comorbidities. In this indication, delta-9-tetrahydrocannabinol (THC)-containing CBMs are recommended. There is limited evidence that CBMs are also effective in Parkinson's disease in which they may improve tremor, but also non-motor symptoms such as pain and sleeping problems. With respect to other neurodegenerative diseases, there is limited evidence that CBMs may improve behavioral symptoms in Huntington's disease. In addition, it has been speculated that CBMs may have neuroprotective effects, but this has not yet been confirmed in the clinical setting.
Collapse
Affiliation(s)
- Natalia Szejko
- Clinic of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Kamila Saramak
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kirsten R Müller-Vahl
- Clinic of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
10
|
Häge A, Krämer R, Dunlap M, Mechler K, Müller-Vahl KR, Nagy P. Emerging therapeutic approaches for Tourette syndrome and other tic disorders - a systematic review of current clinical trials. Eur Child Adolesc Psychiatry 2024:10.1007/s00787-024-02637-x. [PMID: 39714607 DOI: 10.1007/s00787-024-02637-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: 11/07/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Tourette syndrome and other tic disorders are prevalent neurodevelopmental disorders typically treated with behavioral techniques or pharmacological interventions, primarily antipsychotics. However, many patients do not achieve sufficient response to conventional treatments, underscoring the need for further research in this area. To provide a comprehensive overview of ongoing research activities, we systematically searched the clinical registries of the World Health Organization (WHO) and of the United States National Institutes of Health (NIH) for currently planned or ongoing registered clinical studies. Our search identified 21 registered studies, of which seven focus specifically on pharmacological interventions. Potential candidates include the dopamine D1 antagonist ecopipam, the phosphodiesterase inhibitor gemlapodect, and cannabis-based therapies. Additionally, several studies are exploring behavioral interventions including Comprehensive Behavioral Interventions for Tics (CBIT) and mindfulness training, neurostimulation including MRI-navigated transcranial stimulation, theta burst stimulation, and deep brain stimulation, traditional Chinese medicine, and other approaches such as microbiota transplantation. Despite the range of research activities underway, the overall landscape remains limited. In this report we will discuss the findings, current developments, and relevant aspects concerning future research including clinical, scientific, and patient perspectives.
Collapse
Affiliation(s)
- Alexander Häge
- Department of Child & Adolescent Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, J5, Mannheim, 68159, Germany.
| | - Robert Krämer
- Department of Child & Adolescent Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, J5, Mannheim, 68159, Germany
| | - Michele Dunlap
- Tics and Tourette Across the Globe (TTAG), A Global Non-Profit Advocacy Organization, Hannover Medical School, 30625, Hannover, Germany
| | - Konstantin Mechler
- Department of Child & Adolescent Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, J5, Mannheim, 68159, Germany
| | - Kirsten R Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, 30625, Germany
| | - Peter Nagy
- Department of Neurodevelopmental Disorders, Bethesda Children's Hospital, Budapest, Hungary
| |
Collapse
|
11
|
Feng P, Chen Y, Sun K, Wei X, Ding Y, Shang J, Shi Z, Xu X, Guo J, Tian Y. Volatile oil from Acori graminei Rhizoma affected the synaptic plasticity of rats with tic disorders by modulating dopaminergic and glutamatergic systems. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118676. [PMID: 39147000 DOI: 10.1016/j.jep.2024.118676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acori graminei Rhizoma is a commonly used traditional Chinese medicine for treating TD, with its main component being calamus volatile oil. Volatile Oil from Acori graminei Rhizoma (VOA)can protect nerve cells and alleviate learning and memory disorders. However, the mechanism of anti-tic of VOA is still unclear. AIM OF THE STUDY We aimed to explore the effects of Volatile Oil from Acori Tatarinowii Rhizoma (VOA) on striatal dopaminergic and glutamatergic systems and synaptic plasticity of rats with Tic Disorder (TD), as well as its pharmaceutical mechanism against TD. MATERIALS AND METHODS This study involved 48 (three-week-old) Sprague Dawley (SD) rats, which were randomly divided into two primary groups: Control (8) and TD (40). Rats in the TD group were injected intraperitoneally with 3,3-iminodipropionitrile (IDPN) to construct the TD rat model. They were divided into five subgroups: Model, Tiapride, VOA-high, VOA-medium, and VOA-low (N = 8). After modeling, VOA was administrated to rats in the VOA groups through gavage (once/day for four consecutive weeks), while rats in the blank control and model groups received normal saline of the same volume. The animals' behavioral changes were reflected using the stereotypic and motor behavior scores. After interferences, patterns of striatal neurons and the density of dendritic spines were investigated using H&E and Golgi staining, and the ultrastructure of striatal synapses was examined using Transmission Electron Microscopy (TEM). Furthermore, Ca2+ content was determined using the Ca2+ detector, and Dopamine (DA) and Glutamate (GLU) contents in serum and striatum were detected through ELISA. Finally, DRD1, DRD2, AMPAR1, NMPAR1, DAT, VMAT2, CAMKⅡ, and CREB expression in the striatum was detected using Quantitative real-time PCR (qRT-PCR), Western Blotting (WB) and Immunohistochemical (IHC) methods. RESULTS Compared to rats in the blank control and model groups, rats in the VOA groups showed lower stereotypic behavior scores. Furthermore, rats in the VOA groups exhibited relieved, neuron damage and increased quantities of neuronal dendrites and dendritic spines Additionally, based on TEM images show that, the VOA groups showed a clear synaptic structure and increased amounts of postsynaptic dense substances and synaptic vesicles. The VOA groups also exhibited reduced Ca2+ contents, and upregulation of DRD1, DRD2, DAT, AMPAR1, and NMPAR1 and downregulation of VMAT-2, CAMKⅡ, and CREB in the striatum. CONCLUSIONS In summary, VOA could influence synaptic plasticity by tuning the dopaminergic and glutamatergic systems, thus relieving TD.
Collapse
Affiliation(s)
- Peng Feng
- Medical College, Hexi University, Zhangye, Gansu, China.
| | - Yuanhuan Chen
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Kexin Sun
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xing Wei
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanqin Ding
- Paediatrics, Zhangye People's Hospital Affiliated to Hexi University, Zhangye, Gansu, China
| | - Jing Shang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - ZhengGang Shi
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaomin Xu
- Medical College, Hexi University, Zhangye, Gansu, China
| | - Junxiong Guo
- Institute of Traditional Chinese and Western Medicine Integration, Hexi University, Zhangye, Gansu, China
| | - Yongyan Tian
- Silk Road Traditional Chinese Medicine Research Center, Hexi University, Zhangye, Gansu, China
| |
Collapse
|
12
|
Lu MQ, Shi ZG, Shang J, Gao L, Gao L, Gao WJ. ChangPu YuJin Tang improves Tourette disorder symptoms by modulating amino acid neurotransmitters in IDPN model rats. Metab Brain Dis 2024; 39:1543-1558. [PMID: 39312065 DOI: 10.1007/s11011-024-01411-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/09/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Changpu Yujin Tang(CPYJT), a Chinese herbal compound, is an effective therapeutic strategy for pediatric patients with Tourette disorder (TD). Therefore, this work aims to investigate the therapeutic mechanisms of CPYJT. METHODS Behavioral and cellular ultrastructural evaluation of the therapeutic effects of CPYJT in TD model rats. Colorimetric methods, reverse transcription‑quantitative PCR, and Western Blot were used to measure the altered levels of GLU, GABA, and the levels of VGLUT1, GLUD1, GABRA3, and GAD65 in the cortex, striatum, and thalamus of the TD model rats after 7, 14, 21, and 28 days of CPYJT administration. RESULTS CPYJT significantly reduced stereotypic behavior and motor behavior scores in TD model rats. CPYJT ameliorates myelin structural damage in TD model rat neuronal cells. CPYJT decreased GLU content, elevated GABA content, decreased GLUD1 and VGLUT1 levels, and elevated GAD65 and GABRA3 levels in TD model rats' cortex, striatum, and thalamus. CPYJT has different regulatory time points in the cortex, striatum, and thalamus for critical factors of amino acid-based neurotransmission. CONCLUSION CPYJT protects behavioral and structural damage of neuronal cells in multiple brain regions in TD model rats.
Collapse
Affiliation(s)
- Man-Qi Lu
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, shanghai, 200000, China
| | - Zheng-Gang Shi
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China.
| | - Jing Shang
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Lü Gao
- Shanxi University Of Chinese Medicine Third Clinical Medical College Pediatric Teaching and Research Department, Taiyuan, 140100, Shanxi, China
| | - Lei Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Wei-Jiao Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| |
Collapse
|
13
|
Sarchioto M, Howe F, Morgante F, Stern J, Edwards MJ, Martino D. Magnetic resonance spectroscopy reveals evidence of brain oxidative stress in Tourette syndrome. Parkinsonism Relat Disord 2024; 128:107154. [PMID: 39366163 DOI: 10.1016/j.parkreldis.2024.107154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 10/06/2024]
Affiliation(s)
- Marianna Sarchioto
- King's College Hospital NHS Foundation Trust, Neurology Department, London, UK.
| | - Franklyn Howe
- Neuroscience & Cell Biology Research Institute, St George's University of London, London, UK
| | - Francesca Morgante
- Neuroscience & Cell Biology Research Institute, St George's University of London, London, UK
| | - Jeremy Stern
- Atkinson Morley Regional Neuroscience Centre, St George's University of London, London, UK
| | - Mark J Edwards
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Davide Martino
- Department of Clinical Neurosciences, University of Calgary and Hotchkiss Brain Institute, Calgary, AB, Canada
| |
Collapse
|
14
|
Borrego-Ruiz A, Borrego JJ. Neurodevelopmental Disorders Associated with Gut Microbiome Dysbiosis in Children. CHILDREN (BASEL, SWITZERLAND) 2024; 11:796. [PMID: 39062245 PMCID: PMC11275248 DOI: 10.3390/children11070796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
The formation of the human gut microbiome initiates in utero, and its maturation is established during the first 2-3 years of life. Numerous factors alter the composition of the gut microbiome and its functions, including mode of delivery, early onset of breastfeeding, exposure to antibiotics and chemicals, and maternal stress, among others. The gut microbiome-brain axis refers to the interconnection of biological networks that allow bidirectional communication between the gut microbiome and the brain, involving the nervous, endocrine, and immune systems. Evidence suggests that the gut microbiome and its metabolic byproducts are actively implicated in the regulation of the early brain development. Any disturbance during this stage may adversely affect brain functions, resulting in a variety of neurodevelopmental disorders (NDDs). In the present study, we reviewed recent evidence regarding the impact of the gut microbiome on early brain development, alongside its correlation with significant NDDs, such as autism spectrum disorder, attention-deficit/hyperactivity disorder, Tourette syndrome, cerebral palsy, fetal alcohol spectrum disorders, and genetic NDDs (Rett, Down, Angelman, and Turner syndromes). Understanding changes in the gut microbiome in NDDs may provide new chances for their treatment in the future.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, 29071 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, 29010 Málaga, Spain
| |
Collapse
|
15
|
Remahi S, Mabika M, Côté S, Iorio-Morin C, Near J, Hui SCN, Edden RAE, Théoret H, Whittingstall K, Lepage JF. Neurotransmitter levels in the basal ganglia are associated with intracortical circuit activity of the primary motor cortex in healthy humans. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110892. [PMID: 37952692 DOI: 10.1016/j.pnpbp.2023.110892] [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: 06/27/2023] [Revised: 10/10/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND The basal ganglia are strongly connected to the primary motor cortex (M1) and play a crucial role in movement control. Interestingly, several disorders showing abnormal neurotransmitter levels in basal ganglia also present concomitant anomalies in intracortical function within M1. OBJECTIVE/HYPOTHESIS The main aim of this study was to clarify the relationship between neurotransmitter content in the basal ganglia and intracortical function at M1 in healthy individuals. We hypothesized that neurotransmitter content of the basal ganglia would be significant predictors of M1 intracortical function. METHODS We combined magnetic resonance spectroscopy (MRS) and transcranial magnetic stimulation (TMS) to test this hypothesis in 20 healthy adults. An extensive TMS battery probing common measures of intracortical, and corticospinal excitability was administered, and GABA and glutamate-glutamine levels were assessed from voxels placed over the basal ganglia and the occipital cortex (control region). RESULTS Regression models using metabolite concentration as predictor and TMS metrics as outcome measures showed that glutamate level in the basal ganglia significantly predicted short interval intracortical inhibition (SICI) and intracortical facilitation (ICF), while GABA content did not. No model using metabolite measures from the occipital control voxel was significant. CONCLUSIONS Taken together, these results converge with those obtained in clinical populations and suggest that intracortical circuits in human M1 are associated with the neurotransmitter content of connected but distal subcortical structures crucial for motor function.
Collapse
Affiliation(s)
- Sarah Remahi
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Madora Mabika
- University of Galway, School of Medicine, Galway, Ireland
| | - Samantha Côté
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada; Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Christian Iorio-Morin
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Surgery, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Jamie Near
- Physical Sciences Platform, SunnyBrook Health Sciences Center, Toronto, Canada
| | - Steve C N Hui
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Hugo Théoret
- Department of Psychology, Faculty of Arts and Sciences, Université de Montréal, Montréal, Canada
| | - Kevin Whittingstall
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Jean-François Lepage
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada; Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada.
| |
Collapse
|
16
|
Jiang Y, Li Y, Chen X, Zhai R, Peng Y, Tai R, Zhou C, Wang J. Biomarkers and Tourette syndrome: a systematic review and meta-analysis. Front Neurol 2024; 15:1262057. [PMID: 38385037 PMCID: PMC10879287 DOI: 10.3389/fneur.2024.1262057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
OBJECTIVE This research aims to investigate whether peripheral biomarkers might differentiate individuals with Tourette syndrome (TS) from those without the condition. METHODS A broad range of databases was searched through November 2022. This study employed a systematic literature review and subsequent meta-analysis of case-control studies that assessed the aberration of biomarkers of patients with TS and controls. RESULTS A total of 81 studies were identified, out of which 60 met the eligibility criteria for inclusion in the meta-analysis. Following a meticulous screening procedure to determine the feasibility of incorporating case-control studies into the meta-analysis, 13 comparisons were statistically significant [CD3+ T cell, CD4+ T cell, CD4+ T cell to CD8+ T cell ratio, NK-cell, anti-streptolysin O antibodies, anti-DNase antibodies, glutamic acid (Glu), aspartic acid (Asp), ferritin (Fe), zinc (Zn), lead (Pb), vitamin D, and brain-derived neurotrophic factor (BDNF)]. Publication bias was found for anti-streptolysin O antibodies. Suggestive associations were evidenced for norsalsolinol (NSAL), neuron-specific enolase (NSE), and S100B. CONCLUSION In this study, we present empirical evidence substantiating the link between several peripheral biomarkers and the early diagnosis of TS. Larger and more standardized studies are necessary to replicate the observed results, elucidate the specificity of the biomarkers for TS, and evaluate their precision for use in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Junhong Wang
- Department of Pediatrics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Luo L, Zhang L, Huang H, Guan J, Zhang X, Lin Y, Wu R. 3.0 T multi-parametric MRI reveals metabolic and microstructural abnormalities in the posterior visual pathways in patients with thyroid eye disease. Front Neurosci 2024; 17:1306364. [PMID: 38274503 PMCID: PMC10809795 DOI: 10.3389/fnins.2023.1306364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction We aim to explore the microstructural and metabolic changes in visual pathways in patients with thyroid eye disease (TED) using 3T multi-parametric MRI. Methods Thirty-four TED patients (inactive group = 20; active group = 14; acute group = 18; chronic group = 16) and 12 healthy controls (HC) were recruited from November 2020 to July 2021. Proton magnetic resonance spectroscopy (1H-MRS), glutamate chemical exchange saturation transfer (GluCEST) and diffusion kurtosis imaging (DKI) were performed on 3.0T MR scanner. Data analysis and group comparisons were performed after MR data processing. Results As compare to HC group, the levels of total choline (tCh) in optic radiation (OR) in active group ([1.404 ± 0.560] vs. [1.022 ± 0.260]; p < 0.05), together with tCh ([1.415 ± 0.507] vs. [1.022 ± 0.260]; p < 0.05) in OR in acute group were significantly increased. Glutamine (Gln) levels were higher in OR in the chronic group than those in HCs and were positively correlated with the levels of thyroglobulin antibody (TgAb), thyroid peroxidase antibody (TPOAb), free triiodothyronine (FT3) and FT4 in chronic group. Glutamate (Glu) levels by 1H-MRS did not show significant differences between any two groups. Interestingly, MTRasym (3.0 ppm) was higher in OL in inactive group, active group, acute group and chronic group than those in HCs, and was positively correlated with Glu levels in OL in 1H-MRS. Fractional anisotropy (FA) values from DKI in OR in acute group were significantly lower than those in HCs. Discussion Our initial study demonstrate that GluCEST performs better than 1H-MRS to monitor Glu alterations in visual pathway in TED patients. Changes of brain glutamine levels in TED patients are closely related to their associated hormones alterations, indicating that disease injury status could be reflected through non-invasive metabolites detection by brain 1H-MRS. FA is the most sensitive DKI index to reveal the visual pathway impairment in TED patients. Altogether, our study revealed that 3T multiparametric MR techniques are useful to demonstrate metabolic and microstructural alterations in visual pathways in TED patients. We found that damage to visual pathways occurs in mild TED cases, which not only offers a new approach to the diagnosis of dysthyroid optic neuropathy, but also demonstrates neuropathy in TED is a gradual and continuous spatio-emporal progression.
Collapse
Affiliation(s)
- Lan Luo
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
- Radiology Department, Huizhou Central People’s Hospital, Huizhou, Guangdong Province, China
| | - Liling Zhang
- Endocrinology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Huaidong Huang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Jitian Guan
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Xiaolei Zhang
- Radiology Department, Huizhou Central People’s Hospital, Huizhou, Guangdong Province, China
| | - Yan Lin
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Renhua Wu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| |
Collapse
|
18
|
O'Neill J, Diaz MP, Alger JR, Pochon JB, Ghahremani D, Dean AC, Tyndale RF, Petersen N, Marohnic S, Karaiskaki A, London ED. Smoking, tobacco dependence, and neurometabolites in the dorsal anterior cingulate cortex. Mol Psychiatry 2023; 28:4756-4765. [PMID: 37749232 PMCID: PMC10914613 DOI: 10.1038/s41380-023-02247-0] [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: 03/31/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023]
Abstract
Cigarette smoking has a major impact on global health and morbidity, and positron emission tomographic research has provided evidence for reduced inflammation in the human brain associated with cigarette smoking. Given the consequences of inflammatory dysfunction for health, the question of whether cigarette smoking affects neuroinflammation warrants further investigation. The goal of this project therefore was to validate and extend evidence of hypoinflammation related to smoking, and to examine the potential contribution of inflammation to clinical features of smoking. Using magnetic resonance spectroscopy, we measured levels of neurometabolites that are putative neuroinflammatory markers. N-acetyl compounds (N-acetylaspartate + N-acetylaspartylglutamate), glutamate, creatine, choline-compounds (phosphocholine + glycerophosphocholine), and myo-inositol, have all been linked to neuroinflammation, but they have not been examined as such with respect to smoking. We tested whether people who smoke cigarettes have brain levels of these metabolites consistent with decreased neuroinflammation, and whether clinical features of smoking are associated with levels of these metabolites. The dorsal anterior cingulate cortex was chosen as the region-of-interest because of previous evidence linking it to smoking and related states. Fifty-four adults who smoked daily maintained overnight smoking abstinence before testing and were compared with 37 nonsmoking participants. Among the smoking participants, we tested for associations of metabolite levels with tobacco dependence, smoking history, craving, and withdrawal. Levels of N-acetyl compounds and glutamate were higher, whereas levels of creatine and choline compounds were lower in the smoking group as compared with the nonsmoking group. In the smoking group, glutamate and creatine levels correlated negatively with tobacco dependence, and creatine correlated negatively with lifetime smoking, but none of the metabolite levels correlated with craving or withdrawal. The findings indicate a link between smoking and a hypoinflammatory state in the brain, specifically in the dorsal anterior cingulate cortex. Smoking may thereby increase vulnerability to infection and brain injury.
Collapse
Affiliation(s)
- Joseph O'Neill
- Division of Child & Adolescent Psychiatry, Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA
| | - Maylen Perez Diaz
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
- Biogen, Inc., Nashville, TN, USA
| | - Jeffry R Alger
- Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jean-Baptiste Pochon
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Dara Ghahremani
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrew C Dean
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Rachel F Tyndale
- Department of Pharmacology & Toxicology, and Department of Psychiatry, University of Toronto, and Campbell Family Mental Health Research Institute, Centre for Addiction & Mental Health, Toronto, ON, Canada
| | - Nicole Petersen
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Shane Marohnic
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Andrea Karaiskaki
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Edythe D London
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA.
- Jane and Terry Semel Institute for Neuroscience and Human Behavior and the Department of Psychiatry, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Kanaan AS, Yu D, Metere R, Schäfer A, Schlumm T, Bilgic B, Anwander A, Mathews CA, Scharf JM, Müller-Vahl K, Möller HE. Convergent imaging-transcriptomic evidence for disturbed iron homeostasis in Gilles de la Tourette syndrome. Neurobiol Dis 2023; 185:106252. [PMID: 37536382 DOI: 10.1016/j.nbd.2023.106252] [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: 05/10/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023] Open
Abstract
Gilles de la Tourette syndrome (GTS) is a neuropsychiatric movement disorder with reported abnormalities in various neurotransmitter systems. Considering the integral role of iron in neurotransmitter synthesis and transport, it is hypothesized that iron exhibits a role in GTS pathophysiology. As a surrogate measure of brain iron, quantitative susceptibility mapping (QSM) was performed in 28 patients with GTS and 26 matched controls. Significant susceptibility reductions in the patients, consistent with reduced local iron content, were obtained in subcortical regions known to be implicated in GTS. Regression analysis revealed a significant negative association of tic scores and striatal susceptibility. To interrogate genetic mechanisms that may drive these reductions, spatially specific relationships between susceptibility and gene-expression patterns from the Allen Human Brain Atlas were assessed. Correlations in the striatum were enriched for excitatory, inhibitory, and modulatory neurochemical signaling mechanisms in the motor regions, mitochondrial processes driving ATP production and iron‑sulfur cluster biogenesis in the executive subdivision, and phosphorylation-related mechanisms affecting receptor expression and long-term potentiation in the limbic subdivision. This link between susceptibility reductions and normative transcriptional profiles suggests that disruptions in iron regulatory mechanisms are involved in GTS pathophysiology and may lead to pervasive abnormalities in mechanisms regulated by iron-containing enzymes.
Collapse
Affiliation(s)
- Ahmad Seif Kanaan
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.
| | - Dongmei Yu
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Riccardo Metere
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Andreas Schäfer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Siemens Healthcare GmbH, Diagnostic Imaging, Magnetic Resonance, Research and Development, Erlangen, Germany
| | - Torsten Schlumm
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Berkin Bilgic
- Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA; Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alfred Anwander
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Carol A Mathews
- Department of Psychiatry, Center for OCD, Anxiety, and Related Disorders, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jeremiah M Scharf
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
20
|
Kanaan AS, Yu D, Metere R, Schäfer A, Schlumm T, Bilgic B, Anwander A, Mathews CA, Scharf JM, Müller-Vahl K, Möller HE. Convergent imaging-transcriptomic evidence for disturbed iron homeostasis in Gilles de la Tourette syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.15.23289978. [PMID: 37292704 PMCID: PMC10246056 DOI: 10.1101/2023.05.15.23289978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gilles de la Tourette syndrome (GTS) is a neuropsychiatric movement disorder with reported abnormalities in various neurotransmitter systems. Considering the integral role of iron in neurotransmitter synthesis and transport, it is hypothesized that iron exhibits a role in GTS pathophysiology. As a surrogate measure of brain iron, quantitative susceptibility mapping (QSM) was performed in 28 patients with GTS and 26 matched controls. Significant susceptibility reductions in the patient cohort, consistent with reduced local iron content, were obtained in subcortical regions known to be implicated in GTS. Regression analysis revealed a significant negative association of tic scores and striatal susceptibility. To interrogate genetic mechanisms that may drive these reductions, spatially specific relationships between susceptibility and gene-expression patterns extracted from the Allen Human Brain Atlas were assessed. Correlations in the striatum were enriched for excitatory, inhibitory, and modulatory neurochemical signaling mechanisms in the motor regions, mitochondrial processes driving ATP production and iron-sulfur cluster biogenesis in the executive subdivision, and phosphorylation-related mechanisms that affect receptor expression and long-term potentiation. This link between susceptibility reductions and normative transcriptional profiles suggests that disruptions in iron regulatory mechanisms are involved in GTS pathophysiology and may lead to pervasive abnormalities in mechanisms regulated by iron-containing enzymes.
Collapse
Affiliation(s)
- Ahmad Seif Kanaan
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Dongmei Yu
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Riccardo Metere
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Andreas Schäfer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Siemens Healthcare GmbH, Diagnostic Imaging, Magnetic Resonance, Research and Development, Erlangen, Germany
| | - Torsten Schlumm
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Berkin Bilgic
- Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alfred Anwander
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Carol A. Mathews
- Department of Psychiatry, Center for OCD, Anxiety, and Related Disorders, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jeremiah M. Scharf
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Harald E. Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
21
|
Woods DW, Himle MB, Stiede JT, Pitts BX. Behavioral Interventions for Children and Adults with Tic Disorder. Annu Rev Clin Psychol 2023; 19:233-260. [PMID: 37159286 DOI: 10.1146/annurev-clinpsy-080921-074307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Over the past decade, behavioral interventions have become increasingly recognized and recommended as effective first-line therapies for treating individuals with tic disorders. In this article, we describe a basic theoretical and conceptual framework through which the reader can understand the application of these interventions for treating tics. The three primary behavioral interventions for tics with the strongest empirical support (habit reversal, Comprehensive Behavioral Intervention for Tics, and exposure and response prevention) are described. Research on the efficacy and effectiveness of these treatments is summarized along with a discussion of the research evaluating the delivery of these treatments in different formats and modalities. The article closes with a review of the possible mechanisms of change underlying behavioral interventions for tics and areas for future research.
Collapse
Affiliation(s)
- Douglas W Woods
- Department of Psychology, Marquette University, Milwaukee, Wisconsin, USA;
| | - Michael B Himle
- Department of Psychology, University of Utah, Salt Lake City, Utah, USA
| | - Jordan T Stiede
- Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Brandon X Pitts
- Department of Psychology, Marquette University, Milwaukee, Wisconsin, USA;
| |
Collapse
|
22
|
Hao J, Zhang X, Liu Y, Zhang Z, Jiang K, Zhang XY, Wu M. Cross-sectional Exploration of the Relationship Between Glutamate Abnormalities and Tic Disorder Severity Using Proton Magnetic Resonance Spectroscopy. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:138-147. [PMID: 37197641 PMCID: PMC10110806 DOI: 10.1007/s43657-022-00064-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 05/19/2023]
Abstract
Glutamate (Glu) has been reported to be closely related to the pathophysiology of Tic Disorders (TD). By using proton magnetic resonance spectroscopy (1H-MRS), we aimed to investigate the relationship between in vivo Glu levels and the severity of TD. We performed a cross-sectional study in medication-free patients with TD and healthy controls aged between 5 and 13 years using 1H-MRS at 3 T. First, we measured the Glu levels in both patients and controls and observed the difference in subgroups, including mild TD patients and moderate TD patients. We then examined the correlations between the Glu levels and clinical features of the patients. Finally, we assessed the diagnostic value of 1H-MRS and the influencing factors. Our results show that the Glu levels in the striatum of all patients with TD were not significantly different from those of the healthy controls. Subgroup analysis revealed that the Glu levels in the moderate TD group were higher than those in the mild TD group and healthy controls. The correlation analysis showed that Glu levels are strongly positive correlated with TD severity. The optimal cutoff value of Glu levels to differentiate mild tics from moderate tics was 1.244, with a sensitivity of 88.2% and a specificity of 94.7%. Multiple linear regression models revealed that the severity of TD is one of the important factors that affect Glu levels. We conclude that Glu levels are mainly associated with the severity of tics, thus it could serve as a key biomarker for TD classification.
Collapse
Affiliation(s)
- Juanjuan Hao
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 People’s Republic of China
- School of Medicine, Shaoxing University, 900 Chengnan Road, Shaoxing, 312000 People’s Republic of China
| | - Xin Zhang
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 People’s Republic of China
| | - Ying Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433 People’s Republic of China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433 People’s Republic of China
| | - Zhongyang Zhang
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 People’s Republic of China
| | - Keyu Jiang
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 People’s Republic of China
| | - Xiao-Yong Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433 People’s Republic of China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, 200433 People’s Republic of China
| | - Min Wu
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092 People’s Republic of China
| |
Collapse
|
23
|
Molecular Landscape of Tourette's Disorder. Int J Mol Sci 2023; 24:ijms24021428. [PMID: 36674940 PMCID: PMC9865021 DOI: 10.3390/ijms24021428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/12/2023] Open
Abstract
Tourette's disorder (TD) is a highly heritable childhood-onset neurodevelopmental disorder and is caused by a complex interplay of multiple genetic and environmental factors. Yet, the molecular mechanisms underlying the disorder remain largely elusive. In this study, we used the available omics data to compile a list of TD candidate genes, and we subsequently conducted tissue/cell type specificity and functional enrichment analyses of this list. Using genomic data, we also investigated genetic sharing between TD and blood and cerebrospinal fluid (CSF) metabolite levels. Lastly, we built a molecular landscape of TD through integrating the results from these analyses with an extensive literature search to identify the interactions between the TD candidate genes/proteins and metabolites. We found evidence for an enriched expression of the TD candidate genes in four brain regions and the pituitary. The functional enrichment analyses implicated two pathways ('cAMP-mediated signaling' and 'Endocannabinoid Neuronal Synapse Pathway') and multiple biological functions related to brain development and synaptic transmission in TD etiology. Furthermore, we found genetic sharing between TD and the blood and CSF levels of 39 metabolites. The landscape of TD not only provides insights into the (altered) molecular processes that underlie the disease but, through the identification of potential drug targets (such as FLT3, NAALAD2, CX3CL1-CX3CR1, OPRM1, and HRH2), it also yields clues for developing novel TD treatments.
Collapse
|
24
|
Schütteler C, Gerlach AL. Die Bedeutung des Vorgefühls bei Tic-Störungen. ZEITSCHRIFT FUR KLINISCHE PSYCHOLOGIE UND PSYCHOTHERAPIE 2022. [DOI: 10.1026/1616-3443/a000677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Zusammenfassung. Theoretischer Hintergrund: Die Funktion des Vorgefühls in der Pathogenese und Aufrechterhaltung von Tic-Störungen (TS) wird in den letzten Jahren verstärkt erforscht. Die mögliche funktionelle Bedeutung der Vorgefühle wird aber noch nicht ausreichend verstanden. Methode: Im vorliegenden Review wird der Kenntnisstand zu Vorgefühlen entlang eines integrativen funktionalen Störungsmodells zusammengefasst. Ergebnisse: Im Vergleich zum Jugendalter nehmen Tic-Symptome bei Tic-Störungen im Erwachsenenalter ab, während immer mehr Betroffene ein Vorgefühl berichten. Hierbei kann zwischen einem allgemeinen Vorgefühl (trait) und dem Drang, Tics auszuführen (state) unterschieden werden. Das Vorgefühl als trait ist abhängig von der Interozeptionsfähigkeit. An den Drang, Tics auszuführen, kann habituiert werden, moderiert von Aufmerksamkeits- und Attributionsprozessen. Durch das Auflösen des Vorgefühl-Tic-Reizreaktionsmusters reduzieren sich die Tic-Symptome. Schlussfolgerung: Für weitere Erkenntnisse in Bezug auf die Bedeutung von Vorgefühl und den Drang, Tics auszuführen, sollten zukünftige Forschungsansätze Drang und allgemeine Vorgefühle in therapeutischen Interventionsstudien berücksichtigen, weitere Interozeptionsparadigmen einbeziehen und die Entwicklung von allgemeinem Vorgefühl und Drang über die Lebensspanne hinweg untersuchen.
Collapse
Affiliation(s)
- Christina Schütteler
- Institut für Klinische Psychologie und Psychotherapie, Universität zu Köln, Deutschland
| | - Alexander L. Gerlach
- Institut für Klinische Psychologie und Psychotherapie, Universität zu Köln, Deutschland
| |
Collapse
|
25
|
Xi L, Ji X, Ji W, Yang Y, Zhang Y, Long H. Jing-an oral liquid alleviates Tourette syndrome via the NMDAR/MAPK/CREB pathway in vivo and in vitro. PHARMACEUTICAL BIOLOGY 2022; 60:1790-1800. [PMID: 36102587 PMCID: PMC9487928 DOI: 10.1080/13880209.2022.2116056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/01/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Jing-an oral liquid (JA) is a Chinese herbal formula used in the treatment of Tourette syndrome (TS); however, its mechanism is unclear. OBJECTIVE To investigate the effects of JA on amino acid neurotransmitters and microglia activation in vivo and in vitro. MATERIALS AND METHODS Sixty male Sprague-Dawley rats were divided into a control group and 5 TS groups. TS was induced in rats with intraperitoneal injection of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (1 mg/kg) and in BV2 cells with lipopolysaccharide. Control and model rats were administered saline, whereas treatment groups were administered JA (5.18, 10.36, or 20.72 g/kg) or tiapride (a benzamide, 23.5 mg/kg) by gavage once daily for 21 days. Stereotypic behaviour was tested. The levels of N-methyl-d-aspartate receptor (NMDAR)/mitogen-activated protein kinase/cAMP response element-binding protein (CREB)-related proteins in striatum and BV2 cells were measured via western blots. CD11b and IBa1 levels were also measured. Ultra-high-performance liquid-chromatography was used to determine γ-aminobutyric acid (GABA), glutamic acid (Glu), and aspartic acid (ASP) levels. RESULTS JA markedly alleviated the stereotype behaviour (25.92 ± 0.35 to 13.78 ± 0.47) in rats. It also increased NMDAR1 (0.48 ± 0.09 to 0.67 ± 0.08; 0.54 ± 0.07 to 1.19 ± 0.18) expression and down-regulated the expression of p-ERK, p-JNK, p-P38, and p-CREB in BV2 cells and rat striatum. Additionally, Glu, ASP, GABA, CD11b, and IBa1 levels were significantly decreased by JA. DISCUSSION AND CONCLUSIONS JA suppressed microglia activation and regulated the levels of amino acid neurotransmitters, indicating that it could be a promising therapeutic agent for TS.
Collapse
Affiliation(s)
- Leying Xi
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xixi Ji
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxiu Ji
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue’e Yang
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyan Long
- Department of Pediatrics, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric, Nanjing University of Chinese Medicine, Nanjing, China
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
26
|
Gagliano A, Murgia F, Capodiferro AM, Tanca MG, Hendren A, Falqui SG, Aresti M, Comini M, Carucci S, Cocco E, Lorefice L, Roccella M, Vetri L, Sotgiu S, Zuddas A, Atzori L. 1H-NMR-Based Metabolomics in Autism Spectrum Disorder and Pediatric Acute-Onset Neuropsychiatric Syndrome. J Clin Med 2022; 11:6493. [PMID: 36362721 PMCID: PMC9658067 DOI: 10.3390/jcm11216493] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/03/2023] Open
Abstract
We recently described a unique plasma metabolite profile in subjects with pediatric acute-onset neuropsychiatric syndrome (PANS), suggesting pathogenic models involving specific patterns of neurotransmission, neuroinflammation, and oxidative stress. Here, we extend the analysis to a group of patients with autism spectrum disorder (ASD), as a consensus has recently emerged around its immune-mediated pathophysiology with a widespread involvement of brain networks. This observational case-control study enrolled patients referred for PANS and ASD from June 2019 to May 2020, as well as neurotypical age and gender-matched control subjects. Thirty-four PANS outpatients, fifteen ASD outpatients, and twenty-five neurotypical subjects underwent physical and neuropsychiatric evaluations, alongside serum metabolomic analysis with 1H-NMR. In supervised models, the metabolomic profile of ASD was significantly different from controls (p = 0.0001), with skewed concentrations of asparagine, aspartate, betaine, glycine, lactate, glucose, and pyruvate. Metabolomic separation was also observed between PANS and ASD subjects (p = 0.02), with differences in the concentrations of arginine, aspartate, betaine, choline, creatine phosphate, glycine, pyruvate, and tryptophan. We confirmed a unique serum metabolomic profile of PANS compared with both ASD and neurotypical subjects, distinguishing PANS as a pathophysiological entity per se. Tryptophan and glycine appear as neuroinflammatory fingerprints of PANS and ASD, respectively. In particular, a reduction in glycine would primarily affect NMDA-R excitatory tone, overall impairing downstream glutamatergic, dopaminergic, and GABAergic transmissions. Nonetheless, we found metabolomic similarities between PANS and ASD that suggest a putative role of N-methyl-D-aspartate receptor (NMDA-R) dysfunction in both disorders. Metabolomics-based approaches could contribute to the identification of novel ASD and PANS biomarkers.
Collapse
Affiliation(s)
- Antonella Gagliano
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
- Department of Health Science, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Murgia
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Agata Maria Capodiferro
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Marcello Giuseppe Tanca
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Aran Hendren
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Stella Giulia Falqui
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Michela Aresti
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Martina Comini
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Sara Carucci
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Regional Center, ASSL Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, 09126 Cagliari, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Regional Center, ASSL Cagliari, 09126 Cagliari, Italy
| | - Michele Roccella
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90128 Palermo, Italy
| | - Luigi Vetri
- Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Stefano Sotgiu
- Child Neuropsychiatry Unit, Department of Medicine, Surgery and Farmacy, University of Sassari, 07100 Sassari, Italy
| | - Alessandro Zuddas
- Child & Adolescent Neuropsychiatry Unit, Department of Biomedical Sciences, “A. Cao” Paediatric Hospital, University of Cagliari, 09121 Cagliari, Italy
| | - Luigi Atzori
- Clinical Metabolomics Unit, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| |
Collapse
|
27
|
Fan F, Bian Z, Zhang X, Wu H, Wang S, Zhang S, Wang Q, Han F. Big data analytics frameworks for the influence of gut microbiota on the development of tic disorder. Front Comput Neurosci 2022; 16:986591. [PMID: 36093417 PMCID: PMC9452719 DOI: 10.3389/fncom.2022.986591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
The association between gut microbiota and psychiatric disorders has received increasing research attention. Meanwhile, big data analysis has been utilized in many filed including business, human healthcare analysis, etc. The primary objective of this article was to provide insights into Big Data Analytics (BDA) to clarify the association between gut microbiota and TD (Tic disorder). Specifically, we investigated the recent studies related to gut microbiota composition differences in patients with TD compared to health people. We searched on PubMed and Embase (Ovid) databases for relevant published articles until June 15, 2021. A total of 78 TD and 62 health control stool samples were examined. Case-control design was applied in all the studies. No consensus was evident in α-diversity and β-diversity. The abundance of phyla Bacteroidetes and Firmicutes was predominant at the taxa level. Gut microbiota taxonomic differences were found between TD cases and controls, though inconsistently across studies. Further studies are needed to reveal the underlying pathophysiology of TD and correlation between TD and gut microbiota composition.
Collapse
Affiliation(s)
- Fei Fan
- Department of Pediatrics, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Fei Fan,
| | - Zhaoxiang Bian
- Chinese EQUATOR Centre, Hong Kong Chinese Medicine Clinical Study Centre, Chinese Clinical Trial Registry (Hong Kong), School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xuan Zhang
- Chinese EQUATOR Centre, Hong Kong Chinese Medicine Clinical Study Centre, Chinese Clinical Trial Registry (Hong Kong), School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Hongwei Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Simeng Wang
- Department of Pediatrics, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Si Zhang
- Department of Pediatrics, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiong Wang
- Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Fei Han
- Department of Pediatrics, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Fei Han,
| |
Collapse
|
28
|
A Pilot Study on Plasma and Urine Neurotransmitter Levels in Children with Tic Disorders. Brain Sci 2022; 12:brainsci12070880. [PMID: 35884687 PMCID: PMC9313232 DOI: 10.3390/brainsci12070880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Tic disorders (TDs), including Tourette syndrome, are childhood-onset neuropsychiatric disorders characterized by motor and/or vocal tics that commonly affect children’s physical and mental health. The pathogenesis of TDs may be related to abnormal neurotransmitters in the cortico-striatal-thalamo-cortical circuitry, especially dopaminergic, glutamatergic, and serotonergic neurotransmitters. The purpose of this study was to preliminarily investigate the differences in the three types of neurotransmitters in plasma and urine between children with TD and healthy children. Methods: We collected 94 samples of plasma and 69 samples of urine from 3–12-year-old Chinese Han children with TD before treatment. The plasma and urine of the same number of healthy Chinese Han children, matched for age and sex, participating in a physical examination, were collected. Ultra-performance liquid chromatography-tandem mass spectrometry was used to detect the three types of neurotransmitters in the above samples. Results: The plasma levels of norepinephrine, glutamic acid, and γ-aminobutyric acid, and the urine levels of normetanephrine and 5-hydroxyindoleacetic acid were higher in the TD children than in healthy children. The area under the curve (AUC) values of the above neurotransmitters in plasma and urine analyzed by receiver operating characteristic curve analysis were all higher than 0.6, with significant differences. Among them, the combined AUC of dopamine, norepinephrine, normetanephrine, glutamic acid, and γ-aminobutyric acid in the 8–12-year-old subgroup was 0.930, and the sensitivity and specificity for TD were 0.821 and 0.974, respectively (p = 0.000). Conclusions: There are differences in plasma and urine neurotransmitters between TD children and healthy children, which lays a foundation for further research on the pathogenesis of TD.
Collapse
|
29
|
He JL, Mikkelsen M, Huddleston DA, Crocetti D, Cecil KM, Singer HS, Edden RA, Gilbert DL, Mostofsky SH, Puts NA. Frequency and Intensity of Premonitory Urges-to-Tic in Tourette Syndrome Is Associated With Supplementary Motor Area GABA+ Levels. Mov Disord 2022; 37:563-573. [PMID: 34854494 PMCID: PMC9014425 DOI: 10.1002/mds.28868] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/05/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Individuals with Tourette syndrome (TS) often report that they express tics as a means of alleviating the experience of unpleasant sensations. These sensations are perceived as an urge to act and are referred to as premonitory urges. Premonitory urges have been the focus of recent efforts to develop interventions to reduce tic expression in those with TS. OBJECTIVE The aim of this study was to examine the contribution of brain γ-aminobutyric acid (GABA) and glutamate levels of the right primary sensorimotor cortex (SM1), supplementary motor area (SMA), and insular cortex (insula) to tic and urge severity in children with TS. METHODS Edited magnetic resonance spectroscopy was used to assess GABA+ (GABA + macromolecules) and Glx (glutamate + glutamine) of the right SM1, SMA, and insula in 68 children with TS (MAge = 10.59, SDAge = 1.33) and 41 typically developing control subjects (MAge = 10.26, SDAge = 2.21). We first compared GABA+ and Glx levels of these brain regions between groups. We then explored the association between regional GABA+ and Glx levels with urge and tic severity. RESULTS GABA+ and Glx of the right SM1, SMA, and insula were comparable between the children with TS and typically developing control subjects. In children with TS, lower levels of SMA GABA+ were associated with more severe and more frequent premonitory urges. Neither GABA+ nor Glx levels were associated with tic severity. CONCLUSIONS These results broadly support the role of GABAergic neurotransmission within the SMA in the experience of premonitory urges in children with TS. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jason L. He
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - David A. Huddleston
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Deana Crocetti
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Kim M. Cecil
- Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Harvey S. Singer
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, USA,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard A.E. Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Donald L. Gilbert
- Division of Neurology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stewart H. Mostofsky
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, Maryland, USA,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicolaas A.J. Puts
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom,MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom,Correspondence to: Dr. Nicolaas Puts, Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AB, London, United Kingdom;
| |
Collapse
|
30
|
Lai M, Li Y, Luo D, Xu J, Li J. Dopamine-2 receptor antibody encephalitis presenting as pure tongue-biting in a tourette syndrome patient: a case report. BMC Psychiatry 2022; 22:47. [PMID: 35057786 PMCID: PMC8772117 DOI: 10.1186/s12888-021-03683-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 12/29/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Tourette syndrome (TS) is a neuropsychiatric disorder characterized by repetitive and patterned tics. Its onset correlates with dysfunctions in immunological activation and neurotransmitters. Autoimmune movement disorders such as dopamine-2 receptor antibody encephalitis (D2R encephalitis) may go undiagnosed in TS patients seeking medical help for tic symptoms only. Here, we present a clinical case of D2R encephalitis in a TS patient. CASE PRESENTATION A 13-year-old boy with a history of TS presented with acute tongue-biting without positive neurologic examination or auxiliary examination results, except for a weakly positive finding for D2R antibodies in the serum sample. He was initially diagnosed with possible D2R encephalitis, but the influence of TS could not be ruled out. In addition to psychotropics, we administered immunotherapy early based on clinical characteristics, and his symptoms were ameliorated significantly. During the follow-up, he was diagnosed with definite D2R encephalitis, and the dosage of psychotropics was further adjusted for fluctuating symptoms. CONCLUSIONS Our case suggests that clinicians should discern D2R encephalitis in TS patients when tics are the primary symptoms. Administering immunotherapy early, according to clinical characteristics, may benefit the patient. Moreover, the features of premonitory urges could help evaluate the state of TS.
Collapse
Affiliation(s)
- Mingfeng Lai
- grid.13291.380000 0001 0807 1581Mental Health Center West China Hospital, Sichuan University, No. 28 Dian Xin Nan Road, Sichuan, Chengdu 610041 China
| | - Yuanyuan Li
- grid.13291.380000 0001 0807 1581Mental Health Center West China Hospital, Sichuan University, No. 28 Dian Xin Nan Road, Sichuan, Chengdu 610041 China
| | - Dan Luo
- grid.13291.380000 0001 0807 1581Mental Health Center West China Hospital, Sichuan University, No. 28 Dian Xin Nan Road, Sichuan, Chengdu 610041 China
| | - Jiajun Xu
- grid.13291.380000 0001 0807 1581Mental Health Center West China Hospital, Sichuan University, No. 28 Dian Xin Nan Road, Sichuan, Chengdu 610041 China
| | - Jing Li
- Mental Health Center West China Hospital, Sichuan University, No. 28 Dian Xin Nan Road, Sichuan, Chengdu 610041, China.
| |
Collapse
|
31
|
Paschou P, Jin Y, Müller-Vahl K, Möller HE, Rizzo R, Hoekstra PJ, Roessner V, Mol Debes N, Worbe Y, Hartmann A, Mir P, Cath D, Neuner I, Eichele H, Zhang C, Lewandowska K, Munchau A, Verrel J, Musil R, Silk TJ, Hanlon CA, Bihun ED, Brandt V, Dietrich A, Forde N, Ganos C, Greene DJ, Chu C, Grothe MJ, Hershey T, Janik P, Koller JM, Martin-Rodriguez JF, Müller K, Palmucci S, Prato A, Ramkiran S, Saia F, Szejko N, Torrecuso R, Tumer Z, Uhlmann A, Veselinovic T, Wolańczyk T, Zouki JJ, Jain P, Topaloudi A, Kaka M, Yang Z, Drineas P, Thomopoulos SI, White T, Veltman DJ, Schmaal L, Stein DJ, Buitelaar J, Franke B, van den Heuvel O, Jahanshad N, Thompson PM, Black KJ. Enhancing neuroimaging genetics through meta-analysis for Tourette syndrome (ENIGMA-TS): A worldwide platform for collaboration. Front Psychiatry 2022; 13:958688. [PMID: 36072455 PMCID: PMC9443935 DOI: 10.3389/fpsyt.2022.958688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Tourette syndrome (TS) is characterized by multiple motor and vocal tics, and high-comorbidity rates with other neuropsychiatric disorders. Obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), autism spectrum disorders (ASDs), major depressive disorder (MDD), and anxiety disorders (AXDs) are among the most prevalent TS comorbidities. To date, studies on TS brain structure and function have been limited in size with efforts mostly fragmented. This leads to low-statistical power, discordant results due to differences in approaches, and hinders the ability to stratify patients according to clinical parameters and investigate comorbidity patterns. Here, we present the scientific premise, perspectives, and key goals that have motivated the establishment of the Enhancing Neuroimaging Genetics through Meta-Analysis for TS (ENIGMA-TS) working group. The ENIGMA-TS working group is an international collaborative effort bringing together a large network of investigators who aim to understand brain structure and function in TS and dissect the underlying neurobiology that leads to observed comorbidity patterns and clinical heterogeneity. Previously collected TS neuroimaging data will be analyzed jointly and integrated with TS genomic data, as well as equivalently large and already existing studies of highly comorbid OCD, ADHD, ASD, MDD, and AXD. Our work highlights the power of collaborative efforts and transdiagnostic approaches, and points to the existence of different TS subtypes. ENIGMA-TS will offer large-scale, high-powered studies that will lead to important insights toward understanding brain structure and function and genetic effects in TS and related disorders, and the identification of biomarkers that could help inform improved clinical practice.
Collapse
Affiliation(s)
- Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Yin Jin
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Hannover University Medical School, Hannover, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Renata Rizzo
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies, University of Catania, Catania, Italy
| | - Pieter J Hoekstra
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, Netherlands
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, Technische Universität (TU) Dresden, Dresden, Germany
| | - Nanette Mol Debes
- Department of Pediatrics, Herlev University Hospital, Herlev, Denmark
| | - Yulia Worbe
- Department of Neurophysiology, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | | | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Danielle Cath
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, Netherlands
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatic, RWTH Aachen University, Aachen, Germany.,Institute of Neuroscience and Medicine 4, Forschungszentrum Jülich GmbH, Jülich, Germany.,JARA BRAIN-Translational Medicine, Aachen, Germany
| | - Heike Eichele
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Chencheng Zhang
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | | | - Alexander Munchau
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Julius Verrel
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Richard Musil
- Department of Psychiatry and Psychotherapy, Ludwig Maximilians University of Munich, Munich, Germany
| | - Tim J Silk
- Deakin University, Geelong, VIC, Australia
| | - Colleen A Hanlon
- Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Emily D Bihun
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| | - Valerie Brandt
- Centre for Innovation in Mental Health, School of Psychology, University of Southampton, Southampton, United Kingdom
| | - Andrea Dietrich
- University Medical Center Groningen, Department of Psychiatry, University of Groningen, Groningen, Netherlands
| | - Natalie Forde
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, Netherlands
| | - Christos Ganos
- Department of Neurology, Charité-University Medicine Berlin, Berlin, Germany
| | - Deanna J Greene
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, United States
| | - Chunguang Chu
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Tamara Hershey
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| | - Piotr Janik
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Jonathan M Koller
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| | - Juan Francisco Martin-Rodriguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Karsten Müller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Stefano Palmucci
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies, University of Catania, Catania, Italy
| | - Adriana Prato
- Child and Adolescent Neurology and Psychiatric Section, Department of Clinical and Experimental Medicine, Catania University, Catania, Italy
| | - Shukti Ramkiran
- Department of Psychiatry, Psychotherapy and Psychosomatic, RWTH Aachen University, Aachen, Germany.,Institute of Neuroscience and Medicine 4, Forschungszentrum Jülich GmbH, Jülich, Germany.,JARA BRAIN-Translational Medicine, Aachen, Germany
| | - Federica Saia
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, School of Medicine, University of Catania, Catania, Italy
| | - Natalia Szejko
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Renzo Torrecuso
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Zeynep Tumer
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Anne Uhlmann
- Department of Child and Adolescent Psychiatry, Technische Universität (TU) Dresden, Dresden, Germany
| | - Tanja Veselinovic
- Department of Psychiatry, Psychotherapy and Psychosomatic, RWTH Aachen University, Aachen, Germany
| | - Tomasz Wolańczyk
- Department of Child Psychiatry, Medical University of Warsaw, Warsaw, Poland
| | | | - Pritesh Jain
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Apostolia Topaloudi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Mary Kaka
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Zhiyu Yang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Petros Drineas
- Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Sophia I Thomopoulos
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Tonya White
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - Lianne Schmaal
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Dan J Stein
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Jan Buitelaar
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, Netherlands
| | - Barbara Franke
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, Netherlands
| | - Odile van den Heuvel
- Department Psychiatry, Department Anatomy and Neuroscience, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Paul M Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kevin J Black
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
32
|
Chu Q, Song A, Zhao R, Liu J, Shi H, Liu P, Dong C, Yan Z. Establishment and evaluation of a compound fear behavior model of Tourette's syndrome in rats. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1469. [PMID: 34734021 PMCID: PMC8506738 DOI: 10.21037/atm-21-4515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/16/2021] [Indexed: 11/09/2022]
Abstract
Background Tourette syndrome (TS) is a common childhood disorder characterized by unwanted movements or vocal sounds called tics. It is often accompanied by other psychobehavioral disorders, including fearful behavior. The establishment and evaluation of rat models of TS and comorbid fear can provide an experimental basis for the treatment of TS and its comorbid fear disorder. Methods Sixteen rats were randomly divided into a model group (n=8) and control group (n=8). In the model group, rats were injected intraperitoneally with iminodipropionitrile (IDPN) for 1 week to establish the TS model, which was followed by acoustic and electrical stimulation for 3 weeks to establish the rat models of TS and comorbid fear. The control group received intraperitoneal injection of saline for 1 week, and no further intervention was given in the last 3 weeks. The behavioral changes of the rats were observed and analyzed by the open field test (OFT). Protein kinase A (PKA), cyclic adenosine monophosphate (cAMP), and dopamine (DA) were measured by enzyme-linked immunosorbent assay (ELISA), and tyrosine hydroxylase (TH) and microRNA-134 (miRNA-134) in the brain tissue were detected by using polymerase chain reaction (PCR). Results One rat in the model group died on the 24th day. Compared with the control group, the model group had significantly higher scores of locomotor activity, stereotyped behavior, and motor behavior, along with prolonged freezing time and significantly lower expression of miRNA-134. The differences in the expressions of PKA, cAMP, DA, and TH in brain tissue were not statistically significant. Conclusions The rat models of TS and comorbid fear have similar changes in behaviors and miRNA-134 level to those in clinical settings and therefore can be used as a reliable animal model to study the mechanism of action of TS and comorbid fear.
Collapse
Affiliation(s)
- Qian Chu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Anran Song
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Run Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianmin Liu
- Department of Psychosomatic Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huishan Shi
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Pulin Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengda Dong
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaojun Yan
- Department of Psychosomatic Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
33
|
"Glu/GABA-Gln" metabolic loop abnormalities in iminodipropionitrile (IDPN)-induced dyskinetic syndrome. Neurol Sci 2021; 42:4697-4706. [PMID: 34431014 DOI: 10.1007/s10072-021-05570-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Iminodipropionitrile (IDPN)-induced dyskinetic syndrome is characterized by abnormal repetitive involuntary movements with abnormalities in the neuro-transmission. This study explored the mechanism of glutamate (Glu)/γ-aminobutyric acid (GABA)-glutamine (Gln) metabolic circuit in rat dyskinetic syndrome and the possible regulation mechanism of "tiapride (Tia)." METHODS Male Wistar rats were assigned to the control group, dyskinetic syndrome group, and Tia group. Dyskinetic syndrome was induced by injecting with 3,3'-iminodipropionitrile for 7 days. Tia group was treated with tiapride, while the control and dyskinetic syndrome groups were gavaged with saline. Eventually the Glu, GABA, and Gln concentrations in striatum were detected using UPLC-3QMS, additionally another amino acid neurotransmitters (aspartate, glycine) were also detected. Expressions of glutamine synthetase (GS), glutamate transporter (EAAT2), glutamate decarboxylase (GAD65/67), and γ-aminobutyric acid transporter protein (GAD-T) were observed using Western blot and real-time polymerase chain reaction. RESULTS The behavior test scores of dyskinetic syndrome group were increased compared with the control group. Tia group decreased the behavior test scores compared with dyskinetic syndrome group. For amino acid neuro-transmission, dyskinetic syndrome group increased Glu level (p < 0.01), decreased GABA level (p < 0.01), increased Glu/GABA ratio (p < 0.01), and decreased Asp level (p < 0.01) compared with control group. Tia group decreased Glu level (p < 0.01), increased GABA level (p < 0.01), decreased Glu/GABA ratio (p < 0.01), and increased Asp level (p < 0.05) compared with dyskinetic syndrome group. For Glu/GABA-Gln circuit, the protein and mRNA expression of GS and EAAT2 in dyskinetic syndrome group were decreased (p < 0.05). Tia group increased protein and mRNA expression level of GS (p < 0.05) and EAAT2 (p < 0.01). CONCLUSION The rat dyskinetic syndrome has Glu/GABA-Gln abnormalities. "Tiapride" upregulated the protein expression of GS and EAAT2, reduce Glu levels, increase γ-GABA levels, and eventually improve amino acid neurotransmitter imbalance.
Collapse
|
34
|
Molloy EN, Zsido RG, Piecha FA, Beinhölzl N, Scharrer U, Zheleva G, Regenthal R, Sehm B, Nikulin VV, Möller HE, Villringer A, Sacher J, Mueller K. Decreased thalamo-cortico connectivity during an implicit sequence motor learning task and 7 days escitalopram intake. Sci Rep 2021; 11:15060. [PMID: 34301974 PMCID: PMC8302647 DOI: 10.1038/s41598-021-94009-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/05/2021] [Indexed: 11/12/2022] Open
Abstract
Evidence suggests that selective serotonin reuptake inhibitors (SSRIs) reorganize neural networks via a transient window of neuroplasticity. While previous findings support an effect of SSRIs on intrinsic functional connectivity, little is known regarding the influence of SSRI-administration on connectivity during sequence motor learning. To investigate this, we administered 20 mg escitalopram or placebo for 1-week to 60 healthy female participants undergoing concurrent functional magnetic resonance imaging and sequence motor training in a double-blind randomized controlled design. We assessed task-modulated functional connectivity with a psycho-physiological interaction (PPI) analysis in the thalamus, putamen, cerebellum, dorsal premotor, primary motor, supplementary motor, and dorsolateral prefrontal cortices. Comparing an implicit sequence learning condition to a control learning condition, we observed decreased connectivity between the thalamus and bilateral motor regions after 7 days of escitalopram intake. Additionally, we observed a negative correlation between plasma escitalopram levels and PPI connectivity changes, with higher escitalopram levels being associated with greater thalamo-cortico decreases. Our results suggest that escitalopram enhances network-level processing efficiency during sequence motor learning, despite no changes in behaviour. Future studies in more diverse samples, however, with quantitative imaging of neurochemical markers of excitation and inhibition, are necessary to further assess neural responses to escitalopram.
Collapse
Affiliation(s)
- Eóin N Molloy
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany.
- International Max Planck Research School NeuroCom, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Rachel G Zsido
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
- International Max Planck Research School NeuroCom, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Max Planck School of Cognition, Leipzig, Germany
| | - Fabian A Piecha
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
| | - Nathalie Beinhölzl
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
| | - Ulrike Scharrer
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
| | - Gergana Zheleva
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
| | - Ralf Regenthal
- Division of Clinical Pharmacology, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Bernhard Sehm
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
- Department of Neurology, University Hospital Halle (Saale), Halle, Germany
| | - Vadim V Nikulin
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
- Centre for Cognition and Decision Making, Institute for Cognitive Neuroscience, National Research University Higher School of Economics, Moscow, Russia
| | - Harald E Möller
- Nuclear Magnetic Resonance Methods and Development Group, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany
- International Max Planck Research School NeuroCom, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- MindBrainBody Institute, Berlin School of Mind and Brain, Charité-Berlin University of Medicine and Humboldt University Berlin, Berlin, Germany
- Clinic of Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Julia Sacher
- Emotion and Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103, Leipzig, Germany.
- International Max Planck Research School NeuroCom, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
- Max Planck School of Cognition, Leipzig, Germany.
- Clinic of Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany.
| | - Karsten Mueller
- Nuclear Magnetic Resonance Methods and Development Group, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
35
|
The why behind the high: determinants of neurocognition during acute cannabis exposure. Nat Rev Neurosci 2021; 22:439-454. [PMID: 34045693 DOI: 10.1038/s41583-021-00466-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 11/08/2022]
Abstract
Acute cannabis intoxication may induce neurocognitive impairment and is a possible cause of human error, injury and psychological distress. One of the major concerns raised about increasing cannabis legalization and the therapeutic use of cannabis is that it will increase cannabis-related harm. However, the impairing effect of cannabis during intoxication varies among individuals and may not occur in all users. There is evidence that the neurocognitive response to acute cannabis exposure is driven by changes in the activity of the mesocorticolimbic and salience networks, can be exacerbated or mitigated by biological and pharmacological factors, varies with product formulations and frequency of use and can differ between recreational and therapeutic use. It is argued that these determinants of the cannabis-induced neurocognitive state should be taken into account when defining and evaluating levels of cannabis impairment in the legal arena, when prescribing cannabis in therapeutic settings and when informing society about the safe and responsible use of cannabis.
Collapse
|
36
|
Sarchioto M, Howe F, Dumitriu IE, Morgante F, Stern J, Edwards MJ, Martino D. Analyses of peripheral blood dendritic cells and magnetic resonance spectroscopy support dysfunctional neuro-immune crosstalk in Tourette syndrome. Eur J Neurol 2021; 28:1910-1921. [PMID: 33768607 DOI: 10.1111/ene.14837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/27/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Evidence supports that neurodevelopmental diseases, such as Tourette syndrome (TS), may involve dysfunctional neural-immune crosstalk. This could lead to altered brain maturation and differences in immune and stress responses. Dendritic cells (DCs) play a major role in immunity as professional antigen-presenting cells; changes in their frequency have been observed in several autoimmune conditions. METHODS In 18 TS patients (15 on stable pharmacological treatment, three unmedicated) and 18 age-matched healthy volunteers (HVs), we explored circulating blood-derived DCs and their relationship with clinical variables and brain metabolites, measured via proton magnetic resonance spectroscopy (1H-MRS). DC subsets, including plasmacytoid and myeloid type 1 and 2 dendritic cells (MDC1, MDC2), were studied with flow cytometry. 1H-MRS was used to measure total choline, glutamate plus glutamine, total creatine (tCr), and total N-acetylaspartate and N-acetylaspartyl-glutamate levels in frontal white matter (FWM) and the putamen. RESULTS We did not observe differences in absolute concentrations of DC subsets or brain inflammatory metabolites between patients and HVs. However, TS patients manifesting anxiety showed a significant increase in MDC1s compared to TS patients without anxiety (p = 0.01). We also found a strong negative correlation between MDC1 frequency and tCr in the FWM of patients with TS (p = 0.0015), but not of HVs. CONCLUSION Elevated frequencies of the MDC1 subset in TS patients manifesting anxiety may reflect a proinflammatory status, potentially facilitating altered neuro-immune crosstalk. Furthermore, the strong inverse correlation between brain tCr levels and MDC1 subset frequency in TS patients suggests a potential association between proinflammatory status and metabolic changes in sensitive brain regions.
Collapse
Affiliation(s)
- Marianna Sarchioto
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK.,Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Franklyn Howe
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Ingrid E Dumitriu
- Molecular and Clinical Sciences Research Institute, St George's, University of London and Cardiology Clinical Academic Group, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Francesca Morgante
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Jeremy Stern
- Atkinson Morley Regional Neuroscience Centre, St George's University of London, London, UK
| | - Mark J Edwards
- Neurosciences Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Davide Martino
- Department of Clinical Neurosciences, University of Calgary and Hotchkiss Brain Institute, Calgary, AB, Canada
| |
Collapse
|
37
|
O'Rourke T, Martins PT, Asano R, Tachibana RO, Okanoya K, Boeckx C. Capturing the Effects of Domestication on Vocal Learning Complexity. Trends Cogn Sci 2021; 25:462-474. [PMID: 33810982 DOI: 10.1016/j.tics.2021.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 11/28/2022]
Abstract
Domesticated and vocal learning species can serve as informative model organisms for the reduction of reactive aggression and emergence of speech in our lineage. Amidst mounting evidence that domestication modifies vocal repertoires across different species, we focus on the domesticated Bengalese finch, which has a more complex song than the wild-type white-rumped munia. Our explanation for this effect revolves around the glutamate neurotransmitter system. Glutamate signaling (i) is implicated in birdsong learning, (ii) controls dopamine activity in neural circuits crucial for vocal learning, (iii) is disproportionately targeted in the evolution of domesticates, and (iv) regulates stress responses and aggressive behaviors attenuated under domestication. We propose that attenuated excitation of stress-related neural circuits potentiates vocal learning via altered dopaminergic signaling.
Collapse
Affiliation(s)
- Thomas O'Rourke
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain
| | - Pedro Tiago Martins
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain
| | - Rie Asano
- Department of Systematic Musicology, University of Cologne, 50923 Cologne, Germany
| | - Ryosuke O Tachibana
- Center for Evolutionary Cognitive Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Tokyo, Japan
| | - Kazuo Okanoya
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 153-8902 Tokyo, Japan
| | - Cedric Boeckx
- Section of General Linguistics, University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute for Complex Systems (UBICS), 08028 Barcelona, Spain; Catalan Institute for Advanced Studies and Research (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
38
|
Hildonen M, Levy AM, Dahl C, Bjerregaard VA, Birk Møller L, Guldberg P, Debes NM, Tümer Z. Elevated Expression of SLC6A4 Encoding the Serotonin Transporter (SERT) in Gilles de la Tourette Syndrome. Genes (Basel) 2021; 12:86. [PMID: 33445578 PMCID: PMC7827645 DOI: 10.3390/genes12010086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 01/02/2023] Open
Abstract
Gilles de la Tourette syndrome (GTS) is a complex neurodevelopmental disorder characterized by motor and vocal tics. Most of the GTS individuals have comorbid diagnoses, of which obsessive-compulsive disorder (OCD) and attention deficit-hyperactivity disorder (ADHD) are the most common. Several neurotransmitter systems have been implicated in disease pathogenesis, and amongst these, the dopaminergic and the serotonergic pathways are the most widely studied. In this study, we aimed to investigate whether the serotonin transporter (SERT) gene (SLC6A4) was differentially expressed among GTS individuals compared to healthy controls, and whether DNA variants (the SERT-linked polymorphic region 5-HTTLPR, together with the associated rs25531 and rs25532 variants, and the rare Ile425Val variant) or promoter methylation of SLC6A4 were associated with gene expression levels or with the presence of OCD as comorbidity. We observed that SLC6A4 expression is upregulated in GTS individuals compared to controls. Although no specific genotype, allele or haplotype was overrepresented in GTS individuals compared to controls, we observed that the LAC/LAC genotype of the 5-HTTLPR/rs25531/rs25532 three-locus haplotype was associated with higher SLC6A4 mRNA expression levels in GTS individuals, but not in the control group.
Collapse
Affiliation(s)
- Mathis Hildonen
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark; (M.H.); (A.M.L.); (V.A.B.); (L.B.M.)
| | - Amanda M. Levy
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark; (M.H.); (A.M.L.); (V.A.B.); (L.B.M.)
| | - Christina Dahl
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (C.D.); (P.G.)
| | - Victoria A. Bjerregaard
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark; (M.H.); (A.M.L.); (V.A.B.); (L.B.M.)
| | - Lisbeth Birk Møller
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark; (M.H.); (A.M.L.); (V.A.B.); (L.B.M.)
- Institute for Nature, Systems and Models, Roskilde University Center, 4000 Roskilde, Denmark
| | - Per Guldberg
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark; (C.D.); (P.G.)
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Nanette M. Debes
- Tourette Clinics, Department of Paediatrics, Copenhagen University Hospital, 2730 Herlev, Denmark;
| | - Zeynep Tümer
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark; (M.H.); (A.M.L.); (V.A.B.); (L.B.M.)
- Deparment of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2020 Copenhagen, Denmark
| |
Collapse
|
39
|
Xia JD, Chen F, Zhang QJ, Wang YM, Dai YT, Song NH, Wang ZJ, Zhang B, Yang J. Abnormal Thalamic Metabolism in Patients With Lifelong Premature Ejaculation. J Sex Med 2020; 18:275-283. [PMID: 33358559 DOI: 10.1016/j.jsxm.2020.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/22/2020] [Accepted: 11/20/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Although some recent neuroimaging studies have indicated the abnormal brain structure or function in patients with lifelong premature ejaculation (LPE), whether and how the abnormal thalamic function participates in processing sexual behavioral information are still unclear in patients with LPE. AIM The aim of this study was to assess the changes in the thalamus metabolism and structural integrity in patients with LPE. METHODS We performed a multimodal magnetic resonance approach in a 3.0 T system, including proton magnetic resonance spectroscopy (1H-MRS), diffusion tensor imaging, and volumetric analysis to detect the differences in thalamic metabolism and structure between 20 patients with LPE and 15 healthy controls. OUTCOMES We analyzed and correlated the clinical symptoms of the subjects with significant 1H-MRS-based features. Peak areas of N-acetylaspartate, choline, creatine (Cr), and glutamate/glutamine (Glu) were calculated with the LCModel software. RESULTS Diffusion tensor imaging and volumetric analysis of thalami showed no differences between the 2 groups. On the contrary, 1H-MRS study disclosed that both Glu concentrations and Glu/Cr ratio values in the thalami of patients with LPE were remarkably increased when compared with healthy controls (P < .01 for both variables). In addition, both the intravaginal ejaculatory latency time score and Chinese Index of Sexual Function for Premature Ejaculation-5 score were negatively related to increased Glu concentrations and Glu/Cr ratio values. CLINICAL IMPLICATIONS Glutamatergic activity changes of thalamus may be an underlying indicator for evaluating sensory conduction efficiency in patients with LPE. STRENGTHS & LIMITATIONS The present study first found the abnormal thalamic metabolism in patients with LPE and contributed to a better understanding of the LPE etiology. Limitations include a cross-sectional study design with small samples and no examination of other brain areas. CONCLUSION Our findings show that the increase in glutamatergic activity of thalamus is related to LPE, suggesting that the increased Glu neurotransmission in the thalamus may contribute to the development of premature ejaculation. Xia J-D, Chen F, Zhang Q-J, et al. Abnormal Thalamic Metabolism in Patients With Lifelong Premature Ejaculation. J Sex Med 2021;18:275-283.
Collapse
Affiliation(s)
- Jia-Dong Xia
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Fei Chen
- Department of Radiology, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, Jiangsu, China
| | - Qi-Jie Zhang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ya-Min Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu-Tian Dai
- Department of Andrology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Ning-Hong Song
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zeng-Jun Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jie Yang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
40
|
Szejko N, Lombroso A, Bloch MH, Landeros-Weisenberger A, Leckman JF. Refractory Gilles de la Tourette Syndrome-Many Pieces That Define the Puzzle. Front Neurol 2020; 11:589511. [PMID: 33391155 PMCID: PMC7775596 DOI: 10.3389/fneur.2020.589511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/20/2020] [Indexed: 12/27/2022] Open
Abstract
Gilles de la Tourette syndrome (GTS) is a childhood onset neuropsychiatric disorder characterized by the presence of motor and vocal tics. The clinical spectrum of GTS is heterogeneous and varies from mild cases that do not require any medical attention to cases that are refractory to standard treatments. One of the unresolved issues is the definition of what constitutes treatment-refractory GTS. While for some other neuropsychiatric disorders, such as obsessive-compulsive disorder (OCD), a clear definition has been established, there is still no consensus with regard to GTS. One important issue is that many individuals with GTS also meet criteria for one or more other neurodevelopmental and neuropsychiatric disorders. In many individuals, the severity of these comorbid conditions contributes to the degree to which GTS is treatment refractory. The scope of this paper is to present the current state-of-the-art regarding refractory GTS and indicate possible approaches to define it. In closing, we discuss promising approaches to the treatment of individuals with refractory GTS.
Collapse
Affiliation(s)
- Natalia Szejko
- Division of Neurocritical Care & Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT, United States.,Department of Neurology, Medical University of Warsaw, Warsaw, Poland.,Department of Bioethics, Medical University of Warsaw, Warsaw, Poland
| | - Adam Lombroso
- Child Study Center, Departments of Psychiatry, Pediatrics and Psychology, Yale University, New Haven, CT, United States
| | - Michael H Bloch
- Child Study Center, Departments of Psychiatry, Pediatrics and Psychology, Yale University, New Haven, CT, United States
| | - Angeli Landeros-Weisenberger
- Child Study Center, Departments of Psychiatry, Pediatrics and Psychology, Yale University, New Haven, CT, United States
| | - James F Leckman
- Child Study Center, Departments of Psychiatry, Pediatrics and Psychology, Yale University, New Haven, CT, United States
| |
Collapse
|
41
|
Xia X, Lin Y, Lang B, Yuan J, Sheng L, Yang D, Shen J. Characteristics of diffusion tensor imaging of central nervous system in children with tourette's disease. Medicine (Baltimore) 2020; 99:e20492. [PMID: 32481462 DOI: 10.1097/md.0000000000020492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
To investigate the characteristics of diffusion tensor imaging (DTI) of the central nervous system in children with Tourette syndrome (TS).Fifteen children with TS (TS group) and 15 normal children (control group) were studied, and all of them underwent DTI. The apparent diffusion coefficient (ADC) and fractional anisotropy (FA) parameters were calculated using the DTIStudio software. The region of interest was delineated manually. The ADC and FA values of the bilateral caudate nucleus, bilateral globus pallidus, bilateral putamen, bilateral thalamus, and bilateral frontal lobe white matter were measured using the region of interest editor software. The differences of FA values and ADC values between the same brain areas were compared. The associations between ADC, FA values and Yale Global Tic Severity Scale (YGTSS) scores were evaluated by Pearson correlation analyses.The FA values of left globus pallidus and left thalamus were significantly lower in the TS group than in the control group (P < .05), while the ADC values of the right caudate nucleus and bilateral thalamus were significantly higher in the TS group than in the control group (P < .05). The decrease in FA in the left thalamus significantly correlated with the YGTSS score (r = 0.692; P < .05). No correlation was found between FA and ADC values in other brain regions and the YGTSS score (P > .05).After the DTI analyses, abnormalities were found in the left globus pallidus, right caudate nucleus, and bilateral thalamus in children with TS. Especially the changes in the left thalamus structure was crucial in the pathophysiological clock of TS.
Collapse
Affiliation(s)
| | | | - Boxu Lang
- Department of Acupuncture and Massage Rehabilitation, Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, China
| | | | | | | | | |
Collapse
|
42
|
Abstract
Background:Tics, defined as quick, rapid, sudden, recurrent, non-rhythmic motor movements or vocalizations are required components of Tourette Syndrome (TS) - a complex disorder characterized by the presence of fluctuating, chronic motor and vocal tics, and the presence of co-existing neuropsychological problems. Despite many advances, the underlying pathophysiology of tics/TS remains unknown.Objective:To address a variety of controversies surrounding the pathophysiology of TS. More specifically: 1) the configuration of circuits likely involved; 2) the role of inhibitory influences on motor control; 3) the classification of tics as either goal-directed or habitual behaviors; 4) the potential anatomical site of origin, e.g. cortex, striatum, thalamus, cerebellum, or other(s); and 5) the role of specific neurotransmitters (dopamine, glutamate, GABA, and others) as possible mechanisms (Abstract figure).Methods:Existing evidence from current clinical, basic science, and animal model studies are reviewed to provide: 1) an expanded understanding of individual components and the complex integration of the Cortico-Basal Ganglia-Thalamo-Cortical (CBGTC) circuit - the pathway involved with motor control; and 2) scientific data directly addressing each of the aforementioned controversies regarding pathways, inhibition, classification, anatomy, and neurotransmitters.Conclusion:Until a definitive pathophysiological mechanism is identified, one functional approach is to consider that a disruption anywhere within CBGTC circuitry, or a brain region inputting to the motor circuit, can lead to an aberrant message arriving at the primary motor cortex and enabling a tic. Pharmacologic modulation may be therapeutically beneficial, even though it might not be directed toward the primary abnormality.
Collapse
Affiliation(s)
- Harvey S. Singer
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Farhan Augustine
- Department of Neurology, Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
43
|
Martínez-Maestro M, Labadie C, Möller HE. Dynamic metabolic changes in human visual cortex in regions with positive and negative blood oxygenation level-dependent response. J Cereb Blood Flow Metab 2019; 39:2295-2307. [PMID: 30117749 PMCID: PMC6827122 DOI: 10.1177/0271678x18795426] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dynamic metabolic changes were investigated by functional magnetic resonance spectroscopy (fMRS) during sustained stimulation of human primary visual cortex. Two established paradigms, consisting of either a full-field or a small-circle flickering checkerboard, were employed to generate wide-spread areas of positive or negative blood oxygenation level-dependent (BOLD) responses, respectively. Compared to baseline, the glutamate concentration increased by 5.3% (p = 0.007) during activation and decreased by -3.8% (p = 0.017) during deactivation. These changes were positively correlated with the amplitude of the BOLD response (R = 0.60, p = 0.002) and probably reflect changes of tricarboxylic acid cycle activity. During deactivation, the glucose concentration decreased by -7.9% (p = 0.025) presumably suggesting increased consumption or reduced glucose supply. Other findings included an increased concentration of glutathione (4.2%, p = 0.023) during deactivation and a negative correlation of glutathione and BOLD signal changes (R = -0.49, p = 0.012) as well as positive correlations of aspartate (R = 0.44, p = 0.035) and N-acetylaspartylglutamate (R = 0.42, p = 0.035) baseline concentrations with the BOLD response. It remains to be shown in future work if the observed effects on glutamate and glucose levels deviate from the assumption of a direct link between glucose utilization and regulation of blood flow or support previous suggestions that the hemodynamic response is mainly driven by feedforward release of vasoactive messengers.
Collapse
Affiliation(s)
| | - Christian Labadie
- AG Klinische Neuroimmunologie, NeuroCure Clinical Research Center (NCRC), Charité Universitätsmedizin, Berlin, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
44
|
Loo SK, Miyakoshi M, Tung K, Lloyd E, Salgari G, Dillon A, Chang S, Piacentini J, Makeig S. Neural activation and connectivity during cued eye blinks in Chronic Tic Disorders. NEUROIMAGE-CLINICAL 2019; 24:101956. [PMID: 31382238 PMCID: PMC6698693 DOI: 10.1016/j.nicl.2019.101956] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 05/06/2019] [Accepted: 07/20/2019] [Indexed: 12/28/2022]
Abstract
Objective The pathophysiology of Chronic Tic Disorders (CTDs), including Tourette Syndrome, remains poorly understood. The goal of this study was to compare neural activity and connectivity during a voluntary movement (VM) paradigm that involved cued eye blinks among children with and without CTDs. Using the precise temporal resolution of electroencephalography (EEG), we used the timing and location of cortical source resolved spectral power activation and connectivity to map component processes such as visual attention, cue detection, blink regulation and response monitoring. We hypothesized that neural activation and connectivity during the cued eye blink paradigm would be significantly different in regions typically associated with effortful control of eye blinks, such as frontal, premotor, parietal, and occipital cortices between children with and without CTD. Method Participants were 40 children (23 with CTD, 17 age-matched Healthy Control [HC]), between the ages of 8–12 (mean age = 9.5) years old. All participants underwent phenotypic assessment including diagnostic interviews, behavior rating scales and 128-channel EEG recording. Upon presentation of a cue every 3 s, children were instructed to make an exaggerated blink. Results Behaviorally, the groups did not differ in blink number, latency, or ERP amplitude. Within source resolved clusters located in left dorsolateral prefrontal cortex, posterior cingulate, and supplemental motor area, children with CTD exhibited higher gamma band spectral power relative to controls. In addition, significant diagnostic group differences in theta, alpha, and beta band power in inferior parietal cortex emerged. Spectral power differences were significantly associated with clinical characteristics such as tic severity and premonitory urge strength. After calculating dipole density for 76 anatomical regions, the CTD and HC groups had 70% overlap of top regions with the highest dipole density, suggesting that similar cortical networks were used across groups to carry out the VM. The CTD group exhibited significant information flow increase and dysregulation relative to the HC group, particularly from occipital to frontal regions. Conclusion Children with CTD exhibit abnormally high levels of neural activation and dysregulated connectivity among networks used for regulation and effortful control of voluntary eye blinks. First cortical source level EEG study on brain activity and connectivity in CTD. Children with CTD exhibit aberrant levels of neural activation and connectivity. Neural activation was significantly associated with tic severity and premonitory urge.
Collapse
Affiliation(s)
- Sandra K Loo
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America.
| | - Makoto Miyakoshi
- Swartz Center for Neural Computation, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0559, United States of America
| | - Kelly Tung
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - Evan Lloyd
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - Giulia Salgari
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - Andrea Dillon
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - Susanna Chang
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - John Piacentini
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90095, United States of America
| | - Scott Makeig
- Swartz Center for Neural Computation, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0559, United States of America
| |
Collapse
|
45
|
Rae CL, Critchley HD, Seth AK. A Bayesian Account of the Sensory-Motor Interactions Underlying Symptoms of Tourette Syndrome. Front Psychiatry 2019; 10:29. [PMID: 30890965 PMCID: PMC6412155 DOI: 10.3389/fpsyt.2019.00029] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/17/2019] [Indexed: 11/17/2022] Open
Abstract
Tourette syndrome is a hyperkinetic movement disorder. Characteristic features include tics, recurrent movements that are experienced as compulsive and "unwilled"; uncomfortable premonitory sensations that resolve through tic release; and often, the ability to suppress tics temporarily. We demonstrate how these symptoms and features can be understood in terms of aberrant predictive (Bayesian) processing in hierarchical neural systems, explaining specifically: why tics arise, their "unvoluntary" nature, how premonitory sensations emerge, and why tic suppression works-sometimes. In our model, premonitory sensations and tics are generated through over-precise priors for sensation and action within somatomotor regions of the striatum. Abnormally high precision of priors arises through the dysfunctional synaptic integration of cortical inputs. These priors for sensation and action are projected into primary sensory and motor areas, triggering premonitory sensations and tics, which in turn elicit prediction errors for unexpected feelings and movements. We propose experimental paradigms to validate this Bayesian account of tics. Our model integrates behavioural, neuroimaging, and computational approaches to provide mechanistic insight into the pathophysiological basis of Tourette syndrome.
Collapse
Affiliation(s)
- Charlotte L. Rae
- Sackler Centre for Consciousness Science, University of Sussex, Brighton, United Kingdom
- Department of Neuroscience, Brighton and Sussex Medical School, Brighton, United Kingdom
| | - Hugo D. Critchley
- Sackler Centre for Consciousness Science, University of Sussex, Brighton, United Kingdom
- Department of Neuroscience, Brighton and Sussex Medical School, Brighton, United Kingdom
- Sussex Partnership NHS Foundation Trust, Brighton, United Kingdom
| | - Anil K. Seth
- Sackler Centre for Consciousness Science, University of Sussex, Brighton, United Kingdom
- School of Engineering and Informatics, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
46
|
Hao J, Zhang X, Jiang K, Wu M. New insights into the role of neuron-specific enolase in tic disorders. Neurol Sci 2019; 40:1167-1172. [PMID: 30838543 DOI: 10.1007/s10072-019-03811-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 02/26/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Neuron-specific enolase (NSE) has been suggested for demonstrating brain metabolism in neuropsychiatric disorders. This study assessed serum NSE levels in patients with tic disorders (TD). METHODS In this retrospective case-control study, we investigated whether NSE levels were increased in TD patients. Then, the influencing factors and correlations between NSE levels and clinical features were analyzed. Finally, we tested its diagnostic value for identifying tic severity. RESULTS NSE levels were increased in TD patients, although no statistically significant difference was present between transient TD, chronic TD, and Tourette syndrome. Factors influencing NSE levels assessed by multiple linear regression were the Yale Global Tic Severity Scale (YGTSS) global severity scores and gender. There were significant correlations between NSE levels and tic severity. The optimal cut-off value to distinguish mild tics from moderate-severe tics estimated by receiver operating characteristics curve was 24.95 ng/ml (AUC = 0.683). CONCLUSION Our findings suggested that NSE may be a significant biomarker in TD but should be confirmed in further investigation.
Collapse
Affiliation(s)
- Juanjuan Hao
- Shanghai Jiao Tong University School of Medicine, 227 Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Xin Zhang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Keyu Jiang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Min Wu
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
47
|
Tics and stereotypies: A comparative clinical review. Parkinsonism Relat Disord 2019; 59:117-124. [DOI: 10.1016/j.parkreldis.2019.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/28/2018] [Accepted: 02/03/2019] [Indexed: 01/07/2023]
|
48
|
Augustine F, Singer HS. Merging the Pathophysiology and Pharmacotherapy of Tics. Tremor Other Hyperkinet Mov (N Y) 2019; 8:595. [PMID: 30643668 PMCID: PMC6329776 DOI: 10.7916/d8h14jtx] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
Background Anatomically, cortical-basal ganglia-thalamo-cortical (CBGTC) circuits have an essential role in the expression of tics. At the biochemical level, the proper conveyance of messages through these circuits requires several functionally integrated neurotransmitter systems. In this manuscript, evidence supporting proposed pathophysiological abnormalities, both anatomical and chemical is reviewed. In addition, the results of standard and emerging tic-suppressing therapies affecting nine separate neurotransmitter systems are discussed. The goal of this review is to integrate our current understanding of the pathophysiology of Tourette syndrome (TS) with present and proposed pharmacotherapies for tic suppression. Methods For this manuscript, literature searches were conducted for both current basic science and clinical information in PubMed, Google-Scholar, and other scholarly journals to September 2018. Results The precise primary site of abnormality for tics remains undetermined. Although many pathophysiologic hypotheses favor a specific abnormality of the cortex, striatum, or globus pallidus, others recognize essential influences from regions such as the thalamus, cerebellum, brainstem, and ventral striatum. Some prefer an alteration within direct and indirect pathways, whereas others believe this fails to recognize the multiple interactions within and between CBGTC circuits. Although research and clinical evidence supports involvement of the dopaminergic system, additional data emphasizes the potential roles for several other neurotransmitter systems. Discussion A greater understanding of the primary neurochemical defect in TS would be extremely valuable for the development of new tic-suppressing therapies. Nevertheless, recognizing the varied and complex interactions that exist in a multi-neurotransmitter system, successful therapy may not require direct targeting of the primary abnormality.
Collapse
Affiliation(s)
- Farhan Augustine
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harvey S. Singer
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
|
50
|
Hartmann A, Worbe Y. Tourette syndrome: clinical spectrum, mechanisms and personalized treatments. Curr Opin Neurol 2018; 31:504-509. [DOI: 10.1097/wco.0000000000000575] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|