1
|
Zandt MV, Pittenger C. Sexual dimorphism in histamine regulation of striatal dopamine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595049. [PMID: 38826392 PMCID: PMC11142073 DOI: 10.1101/2024.05.20.595049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Many neuropsychiatric disorders show sex differences in prevalence and presentation. For example, Tourette's Syndrome (TS) is diagnosed 3-5 times more often in males. Dopamine modulation of the basal ganglia is implicated in numerous neuropsychiatric conditions, including TS. Motivated by an unexpected genetic finding in a family with TS, we previously characterized the modulation of striatal dopamine by histamine. Methods We used microdialysis to analyze striatal dopamine response to the targeted infusion of histamine and histamine agonists. siRNA knockdown of histamine receptors was used to identify the cellular mediators of observed effects. Results Intracerebroventricular histamine reduced striatal dopamine in male mice, replicating previous work. Unexpectedly, histamine increased striatal dopamine in females. Targeted infusion of selected agonists revealed that the effect in males depends on H2R receptors in the substantia nigra pars compacta (SNc). Knockdown of H2R in SNc GABAergic neurons abrogated the effect, identifying these cells as a key locus of histamine's regulation of dopamine in males. In females, in contrast, H2R had no role; instead, H3R agonists in the striatum increased striatal dopamine. Strikingly, the effect of histamine on dopamine in females was modulated by the estrous cycle, appearing in estrus/proestrus but not in metestrus/diestrus. Conclusions These findings confirm the regulation of striatal dopamine by histamine but identify marked sexual dimorphism in and estrous modulation of this effect. These findings may shed light on the mechanistic underpinnings of other sex differences in the striatal circuitry, perhaps including the marked sex differences seen in TS and related neuropsychiatric conditions.
Collapse
Affiliation(s)
- Meghan Van Zandt
- Pittenger Laboratory, Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA
| | - Christopher Pittenger
- Pittenger Laboratory, Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale School of Arts and Sciences, New Haven, USA
- Center for Brain and Mind Health, Yale University School of Medicine, New Haven, USA
- Wu-Tsai Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
2
|
Ikrin AN, Moskalenko AM, Mukhamadeev RR, de Abreu MS, Kolesnikova TO, Kalueff AV. The emerging complexity of molecular pathways implicated in mouse self-grooming behavior. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110840. [PMID: 37580009 DOI: 10.1016/j.pnpbp.2023.110840] [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: 06/19/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Rodent self-grooming is an important complex behavior, and its deficits are translationally relevant to a wide range of neuropsychiatric disorders. Here, we analyzed a comprehensive dataset of 227 genes whose mutations are known to evoke aberrant self-grooming in mice. Using these genes, we constructed the network of their established protein-protein interactions (PPI), yielding several distinct molecular clusters related to postsynaptic density, the Wnt signaling, transcription factors, neuronal cell cycle, NOS neurotransmission, microtubule regulation, neuronal differentiation/trafficking, neurodevelopment and mitochondrial function. Utilizing further bioinformatics analyses, we also identified novel central ('hub') proteins within these clusters, whose genes may also be implicated in aberrant self-grooming and other repetitive behaviors in general. Untangling complex molecular pathways of this important behavior using in silico approaches contributes to our understanding of related neurological disorders, and may suggest novel potential targets for their pharmacological or gene therapy.
Collapse
Affiliation(s)
- Aleksey N Ikrin
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Anastasia M Moskalenko
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Radmir R Mukhamadeev
- Graduate Program in Bioinformatics and Genomics, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Murilo S de Abreu
- Moscow Institute of Science and Technology, Dolgoprudny 197028, Russia.
| | - Tatiana O Kolesnikova
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Allan V Kalueff
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 194021, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny 197758, Russia; Neuroscience Group, Ural Federal University, Ekaterinburg 620002, Russia; Laboratory of Translational Biopsychiatry, Scientific Research Institute of Neurosciences and Medicine, Novosibirsk 630117, Russia.
| |
Collapse
|
3
|
Eissa N, Awad MA, Thomas SD, Venkatachalam K, Jayaprakash P, Zhong S, Stark H, Sadek B. Simultaneous Antagonism at H3R/D2R/D3R Reduces Autism-like Self-Grooming and Aggressive Behaviors by Mitigating MAPK Activation in Mice. Int J Mol Sci 2022; 24:ijms24010526. [PMID: 36613969 PMCID: PMC9820264 DOI: 10.3390/ijms24010526] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Dysregulation in brain neurotransmitters underlies several neuropsychiatric disorders, e.g., autism spectrum disorder (ASD). Also, abnormalities in the extracellular-signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway pave the way for neuroinflammation, neurodegeneration, and altered learning phenotype in ASD. Therefore, the effects of chronic systemic administration of the multiple-targeting antagonist ST-713 at the histamine H3 receptor (H3R) and dopamine D2/D3 receptors (D2/D3R) on repetitive self-grooming, aggressive behaviors, and abnormalities in the MAPK pathway in BTBR T + Itpr3tf/J (BTBR) mice were assessed. The results showed that ST-713 (2.5, 5, and 10 mg/kg, i.p.) mitigated repetitive self-grooming and aggression in BTBR mice (all p < 0.05), and the ameliorative effects of the most promising dose of ST-713 (5 mg/kg, i.p.) on behaviors were completely abrogated by co-administration of the H3R agonist (R)-α-methylhistamine or the anticholinergic drug scopolamine. Moreover, the elevated levels of several MAPK pathway proteins and induced proinflammatory markers such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and IL-6 were significantly suppressed following chronic administration of ST-713 (5 mg/kg, i.p.) (all p < 0.01). Furthermore, ST-713 significantly increased the levels of histamine and dopamine in hippocampal tissue of treated BTBR mice (all p < 0.01). The current observations signify the potential role of such multiple-targeting compounds, e.g., ST-713, in multifactorial neurodevelopmental disorders such as ASD.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Mohamed Al Awad
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Karthikkumar Venkatachalam
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sicheng Zhong
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Correspondence:
| |
Collapse
|
4
|
Ramteke A, Lamture Y. Tics and Tourette Syndrome: A Literature Review of Etiological, Clinical, and Pathophysiological Aspects. Cureus 2022; 14:e28575. [PMID: 36185878 PMCID: PMC9520955 DOI: 10.7759/cureus.28575] [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: 08/02/2022] [Accepted: 08/30/2022] [Indexed: 11/07/2022] Open
Abstract
Tourette syndrome (TS) is a condition characterized by tics produced because of neuropsychiatric malfunctioning occurring in childhood, which becomes less severe in adulthood, followed by a difference in the severity of tics between two persons. TS is a diverse variable in which symptoms vary in different patients. It is associated with comorbidities like obsessive-compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), and depression, and hampers the quality of life. Comorbid disorders must be investigated and treated as part of the clinical approach for all TS patients. Clinicians should be aware of the infrequent but serious neurological problems that can occur in these patients and recommend aggressively treating tics. Currently, there is more emphasis on symptom-based treatments by medicines, but as etiological knowledge improves, we will divert to disease-modifying medications in the future. Behavioral, pharmacological, and surgical methods can treat TS. Neuroleptics, other drugs, and behavioral therapies are the first-line options. Deep brain stimulation is evolving but has its pros and cons. The main focus of this review is on tics characteristics, how to manage and assess them, and limitations in the clinical spectrum.
Collapse
|
5
|
Oral histidine intake improves working memory through the activation of histaminergic nervous system in mice. Biochem Biophys Res Commun 2022; 609:141-148. [DOI: 10.1016/j.bbrc.2022.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/05/2022] [Indexed: 11/23/2022]
|
6
|
Xu L, Zhang C, Zhong M, Che F, Guan C, Zheng X, Liu S. Role of histidine decarboxylase gene in the pathogenesis of Tourette syndrome. Brain Behav 2022; 12:e2511. [PMID: 35114079 PMCID: PMC8933785 DOI: 10.1002/brb3.2511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/18/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Tourette syndrome (TS) is caused by complex genetic and environmental factors and is characterized by tics. Histidine decarboxylase (HDC) mutation is a rare genetic cause with high penetrance in patients with TS. HDC-knockout (KO) mice have similar behavioral and neurochemical abnormalities as patients with TS. Therefore, HDC-KO mice are considered a valuable TS pathophysiological model as it reveals the underlying pathological mechanisms that cannot be obtained from patients with TS, thus advancing the development of treatment strategies for TS and other tic disorders. This review summarizes some of the recent research hotspots and progress in HDC-KO mice, aiming to deepen our understanding of brain mechanisms relevant to TS. Furthermore, we encapsulate the possible brain nerve cell changes in HDC-KO mice and their potential roles in TS to provide multiple directions for the future research on tics.
Collapse
Affiliation(s)
- Lulu Xu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Zhang
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, Shandong, China
| | - Meixiang Zhong
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengyuan Che
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, Shandong, China
| | - Chengcheng Guan
- Department of Medical Cenetics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xueping Zheng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shiguo Liu
- Department of Medical Cenetics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
7
|
Pittenger C, Brennan BP, Koran L, Mathews CA, Nestadt G, Pato M, Phillips KA, Rodriguez CI, Simpson HB, Skapinakis P, Stein DJ, Storch EA. Specialty knowledge and competency standards for pharmacotherapy for adult obsessive-compulsive disorder. Psychiatry Res 2021; 300:113853. [PMID: 33975093 PMCID: PMC8536398 DOI: 10.1016/j.psychres.2021.113853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Obsessive-compulsive disorder (OCD) affects approximately one person in 40 and causes substantial suffering. Evidence-based treatments can benefit many; however, optimal treatment can be difficult to access. Diagnosis is frequently delayed, and pharmacological and psychotherapeutic interventions often fail to follow evidence-based guidelines. To ameliorate this distressing situation, the International OCD Accreditation Task Force of the Canadian Institute for Obsessive-Compulsive Disorders has developed knowledge and competency standards for specialized treatments for OCD through the lifespan. These are foundational to evidence-based practice and will form the basis for upcoming ATF development of certification/accreditation programs. Here, we present specialty standards for the pharmacological treatment of adult OCD. We emphasize the importance of integrating pharmacotherapy with clear diagnosis, appreciation of complicating factors, and evidence-based cognitive behavioral therapy. Clear evidence exists to inform first- and second-line pharmacological treatments. In disease refractory to these initial efforts, multiple strategies have been investigated, but the evidence is more equivocal. These standards summarize this limited evidence to give the specialist practitioner a solid basis on which to make difficult decisions in complex cases. It is hoped that further research will lead to development of a clear, multi-step treatment algorithm to support each step in clinical decision-making.
Collapse
Affiliation(s)
- Christopher Pittenger
- Department of Psychiatry and Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States.
| | - Brian P Brennan
- Biological Psychiatry Laboratory and Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Lorrin Koran
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Carol A Mathews
- Department of Psychiatry, University of Florida, Gainesville, FL, United States
| | - Gerald Nestadt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michele Pato
- Institute for Genomic Health and Department of Psychiatry, SUNY Downstate College of Medicine, Brooklyn, NY, United States
| | - Katharine A Phillips
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, and Department of Psychiatry, Weill Cornell Medical College, New York, NY, United States
| | - Carolyn I Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - H Blair Simpson
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States; Office of Mental Health, Research Foundation for Mental Hygiene, New York Psychiatric Institute, New York, NY, United States
| | - Petros Skapinakis
- Department of Psychiatry, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Eric A Storch
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
8
|
Venkatachalam K, Eissa N, Awad MA, Jayaprakash P, Zhong S, Stölting F, Stark H, Sadek B. The histamine H3R and dopamine D2R/D3R antagonist ST-713 ameliorates autism-like behavioral features in BTBR T+tf/J mice by multiple actions. Biomed Pharmacother 2021; 138:111517. [PMID: 33773463 DOI: 10.1016/j.biopha.2021.111517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/07/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022] Open
Abstract
Several brain neurotransmitters, including histamine (HA), acetylcholine (ACh), and dopamine (DA) are suggested to be involved in several brain disorders including cognitive deficits, depression, schizophrenia, anxiety, and narcolepsy, all of which are comorbid with Autism spectrum disorder (ASD). Therefore, the ameliorative effects of the novel multiple-active compound ST-713 with high binding affinities at histamine H3 receptor (H3R), dopamine D2sR and D3R on ASD-like behaviors in male BTBR T+tf/J mice model were assessed. ST-713 (3-(2-chloro-10H-phenothiazin-10-yl)-N-methyl-N-(4-(3-(piperidin-1-yl)propoxy)benzyl)propan-1-amine; 2.5, 5, and 10 mg/kg, i.p.) ameliorated dose-dependently social deficits, and significantly alleviated the repetitive/compulsive behaviors of BTBR mice (all P < 0.05). Moreover, ST-713 modulated disturbed anxiety levels, but failed to obliterate increased hyperactivity of tested mice. Furthermore, ST-713 (5 mg/kg) attenuated the increased levels of hippocampal and cerebellar protein expressions of NF-κB p65, COX-2, and iNOS in BTBR mice (all P < 0.05). The ameliorative effects of ST-713 on social parameters were entirely reversed by co-administration of the H3R agonist (R)-α-methylhistamine or the anticholinergic drug scopolamine. The obtained results demonstrate the potential of multiple-active compounds for the therapeutic management of neuropsychiatric disorders, e.g. ASD.
Collapse
Affiliation(s)
- Karthikkumar Venkatachalam
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Nermin Eissa
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Department of Applied Sciences, College of Arts and Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - Mohamed Al Awad
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates
| | - Sicheng Zhong
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Frauke Stölting
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, 40225 Düsseldorf, Germany
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
9
|
Hsu CJ, Wong LC, Lee WT. Immunological Dysfunction in Tourette Syndrome and Related Disorders. Int J Mol Sci 2021; 22:ijms22020853. [PMID: 33467014 PMCID: PMC7839977 DOI: 10.3390/ijms22020853] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/28/2022] Open
Abstract
Chronic tic disorder and Tourette syndrome are common childhood-onset neurological diseases. However, the pathophysiology underlying these disorders is unclear, and most studies have focused on the disinhibition of the corticostriatal–thalamocortical circuit. An autoimmune dysfunction has been proposed in the pathogenetic mechanism of Tourette syndrome and related neuropsychiatric disorders such as obsessive–compulsive disorder, autism, and attention-deficit/hyperactivity disorder. This is based on evidence from animal model studies and clinical findings. Herein, we review and give an update on the clinical characteristics, clinical evidence, and genetic studies in vitro as well as animal studies regarding immune dysfunction in Tourette syndrome.
Collapse
Affiliation(s)
- Chia-Jui Hsu
- Department of Pediatrics, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 300, Taiwan;
| | - Lee-Chin Wong
- Department of Pediatrics, Cathay General Hospital, Taipei 106, Taiwan;
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Wang-Tso Lee
- Department of Pediatric Neurology, National Taiwan University Children’s Hospital, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 71545); Fax: +886-2-2314-7450
| |
Collapse
|
10
|
Abstract
Effective pharmacological and psychotherapeutic treatments are well established for obsessive-compulsive disorder (OCD). Serotonin reuptake inhibitors (SRIs) are first-line treatment and are of benefit to about half of patients. Augmentation of SRI treatment with low-dose neuroleptics is an evidence-based second-line strategy. Specialty psychotherapy is also used as both first-line and second-line treatment and can benefit many. However, a substantial number of patients do not respond to these treatments. New alternatives are urgently needed. This review summarizes evidence for these established pharmacotherapeutic strategies, and for others that have been investigated in refractory disease but are not supported by the same level of evidence. We focus on three neurotransmitter systems in the brain: serotonin, dopamine, and glutamate. We summarize evidence from genetic, neuroimaging, animal model, and other lines of investigation that probe these three systems in patients with OCD. We also review recent work on predictors of response to current treatments. While many studies suggest abnormalities that may provide insight into the pathophysiology of the disorder, most studies have been small, and non-replication of reported findings has been common. Nevertheless, the gradual accrual of evidence for neurotransmitter dysregulation may in time lead the way to new pharmacological strategies.
Collapse
|
11
|
Zhang C, Xu L, Zheng X, Liu S, Che F. Role of Ash1l in Tourette syndrome and other neurodevelopmental disorders. Dev Neurobiol 2020; 81:79-91. [PMID: 33258273 PMCID: PMC8048680 DOI: 10.1002/dneu.22795] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023]
Abstract
Ash1l potentially contributes to neurodevelopmental diseases. Although specific Ash1l mutations are rare, they have led to informative studies in animal models that may bring therapeutic advances. Ash1l is highly expressed in the brain and correlates with the neuropathology of Tourette syndrome (TS), autism spectrum disorder, and intellectual disability during development, implicating shared epigenetic factors and overlapping neuropathological mechanisms. Functional convergence of Ash1l generated several significant signaling pathways: chromatin remodeling and transcriptional regulation, protein synthesis and cellular metabolism, and synapse development and function. Here, we systematically review the literature on Ash1l, including its discovery, expression, function, regulation, implication in the nervous system, signaling pathway, mutations, and putative involvement in TS and other neurodevelopmental traits. Such findings highlight Ash1l pleiotropy and the necessity of transcending a single gene to complicated mechanisms of network convergence underlying these diseases. With the progress in functional genomic analysis (highlighted in this review), and although the importance and necessity of Ash1l becomes increasingly apparent in the medical field, further research is required to discover the precise function and molecular regulatory mechanisms related to Ash1l. Thus, a new perspective is proposed for basic scientific research and clinical interventions for cross‐disorder diseases.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, China
| | - Lulu Xu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueping Zheng
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fengyuan Che
- Department of Neurology, The Eleventh Clinical Medical College of Qingdao University, Linyi People's Hospital, Linyi, China
| |
Collapse
|
12
|
Berteotti C, Lo Martire V, Alvente S, Bastianini S, Matteoli G, Ohtsu H, Lin JS, Silvani A, Zoccoli G. Tibialis anterior electromyographic bursts during sleep in histamine-deficient mice. J Sleep Res 2020; 30:e13255. [PMID: 33314463 DOI: 10.1111/jsr.13255] [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: 06/06/2020] [Revised: 10/02/2020] [Accepted: 11/18/2020] [Indexed: 11/30/2022]
Abstract
Antihistamine medications have been suggested to elicit clinical features of restless legs syndrome. The available data are limited, particularly concerning periodic leg movements during sleep, which are common in restless legs syndrome and involve bursts of tibialis anterior electromyogram. Here, we tested whether the occurrence of tibialis anterior electromyogram bursts during non-rapid eye movement sleep is altered in histidine decarboxylase knockout mice with congenital histamine deficiency compared with that in wild-type control mice. We implanted six histidine decarboxylase knockout and nine wild-type mice to record neck muscle electromyogram, bilateral tibialis anterior electromyogram, and electroencephalogram during the rest (light) period. The histidine decarboxylase knockout and wild-type mice did not differ significantly in terms of sleep architecture. In both histidine decarboxylase knockout and wild-type mice, the distribution of intervals between tibialis anterior electromyogram bursts had a single peak for intervals < 10 s. The total occurrence rate of tibialis anterior electromyogram bursts during non-rapid eye movement sleep and the occurrence rate of the tibialis anterior electromyogram bursts separated by intervals < 10 s were significantly lower in histidine decarboxylase knockout than in wild-type mice. These data do not support the hypothesis that preventing brain histamine signalling may promote restless legs syndrome. Rather, the data suggest that limb movements during sleep, including those separated by short intervals, are a manifestation of subcortical arousal requiring the integrity of brain histamine signalling.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Viviana Lo Martire
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Sara Alvente
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Stefano Bastianini
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Gabriele Matteoli
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | | | - Jian-Sheng Lin
- Physiologie intégrée du système d'éveil, Centre de recherche en neurosciences de Lyon, INSERM U1028-CNRS UMR 5292 Faculté de Médecine, Université Claude Bernard, Lyon, France
| | - Alessandro Silvani
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum, Università di Bologna, Bologna, Italy
| |
Collapse
|
13
|
Ningdong Granule Upregulates the Striatal DA Transporter and Attenuates Stereotyped Behavior of Tourette Syndrome in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2980705. [PMID: 33005197 PMCID: PMC7509575 DOI: 10.1155/2020/2980705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/15/2020] [Accepted: 08/28/2020] [Indexed: 12/01/2022]
Abstract
This study aimed to evaluate the possible mechanism of Ningdong granule (NDG) for the treatment of Tourette syndrome (TS). The rats with stereotyped behavior were established by microinjection with TS patients' sera; then, the model rats were divided into NDG and haloperidol (Hal) group, and the nonmedication model rats were regarded as treatment control (TS group). The stereotyped behavior of the rats was recorded, the level of dopamine (DA) in striatum, and the content of homovanillic acid (HVA) in sera were tested, and dopamine transporter (DAT) expression was measured in the study. The experimental results showed that NDG effectively inhibited the stereotyped behavior (P < 0.01), decreased the levels of DA in the striatum (P < 0.05), increased the content of sera HVA (P < 0.01), and enhanced the protein and mRNA expression of DAT in the striatum (P < 0.01). Additionally, the results also revealed Hal could improve the stereotyped behavior as well but had no remarkable influence on DAT expression and DA metabolism. In conclusion, NDG attenuates stereotyped behavior, and its mechanism of action might be associated with the upregulation of DAT expression to regulate DA metabolism in the brain.
Collapse
|
14
|
Yoshikawa T, Nakamura T, Yanai K. Histaminergic neurons in the tuberomammillary nucleus as a control centre for wakefulness. Br J Pharmacol 2020; 178:750-769. [PMID: 32744724 DOI: 10.1111/bph.15220] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022] Open
Abstract
Histamine plays pleiotropic roles as a neurotransmitter in the physiology of brain function, this includes the maintenance of wakefulness, appetite regulation and memory retrieval. Since numerous studies have revealed an association between histaminergic dysfunction and diverse neuropsychiatric disorders, such as Alzheimer's disease and schizophrenia, a large number of compounds acting on the brain histamine system have been developed to treat neurological disorders. In 2016, pitolisant, which was developed as a histamine H3 receptor inverse agonist by Schwartz and colleagues, was launched for the treatment of narcolepsy, emphasising the prominent role of brain histamine on wakefulness. Recent advances in neuroscientific techniques such as chemogenetic and optogenetic approaches have led to remarkable progress in the understanding of histaminergic neural circuits essential for the control of wakefulness. In this review article, we summarise the basic knowledge about the histaminergic nervous system and the mechanisms underlying sleep/wake regulation that are controlled by the brain histamine system. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
Collapse
Affiliation(s)
- Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadaho Nakamura
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
15
|
Zhao L, Cheng N, Sun B, Wang S, Li A, Wang Z, Wang Y, Qi F. Regulatory effects of Ningdong granule on microglia-mediated neuroinflammation in a rat model of Tourette's syndrome. Biosci Trends 2020; 14:271-278. [PMID: 32741856 DOI: 10.5582/bst.2020.03262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tourette's syndrome (TS) is an inherited neurologic disorder characterized by involuntary stereotyped motor and vocal tics. Its pathogenesis is still unclear and its treatment remains limited. Recent research has suggested the involvement of immune mechanisms in the pathophysiology of TS. Microglia are the brain's resident innate immune cells. They can mediate neuroinflammation and regulate brain development and homeostasis. A traditional Chinese medicine (TCM), Ningdong granule (NDG), has been found to be efficacious in the treatment of TS while causing few adverse reactions. In the current study, a rat model of 3,3'-iminodipropionitrile (IDPN)-induced TS was used to explore the regulating effects and mechanisms of NDG on microglia-mediated neuroinflammation. IDNP led to robust pathological changes and neurobehavioral complications, with activation of microglia in the striatum of rats with TS. After activation by IDNP, microglia strongly responded to this specific injury, and TNF-α, IL-6, and MCP-1 were released in the striatum and/or serum of rats with TS. Interestingly, NDG inhibited the activation of microglia and decreased the abnormal expression of TNF-α, IL-6, and MCP-1 in the striatum and/or serum of rats with TS, thus controlling tics. However, there were no significant changes in the striatum and/or serum of rats with TS after treatment with haloperidol. The anti-TS action of haloperidol might occur not through microglial activation and neuroinflammation but through the DAT system, thus controlling tics. In conclusion, microglia might play key roles in mediating neuroinflammatory responses in TS, triggering the release of TNF-α, IL-6, and MCP-1.NDG inhibited tics in rats with TS, and this mechanism may be associated with a reduction in the increased number of activated microglia and a decrease in the expression of pro-inflammatory cytokines and chemokines in the striatum and/or serum.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| | - Nan Cheng
- Department of Intensive Care Unit, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| | - Bo Sun
- Shandong Medical Imaging Research Institute affiliated to Shandong University, Ji'nan, China
| | - Shuzhen Wang
- Department of Pediatry, The First Affiliated Hospital of Shandong First Medical University, Ji'nan, China
| | - Anyuan Li
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| | - Zhixue Wang
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| | - Yuan Wang
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| | - Fanghua Qi
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University, Ji'nan, China
| |
Collapse
|
16
|
Biliary damage and liver fibrosis are ameliorated in a novel mouse model lacking l-histidine decarboxylase/histamine signaling. J Transl Med 2020; 100:837-848. [PMID: 32054995 PMCID: PMC7286781 DOI: 10.1038/s41374-020-0405-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/15/2020] [Accepted: 01/18/2020] [Indexed: 02/07/2023] Open
Abstract
Primary sclerosing cholangitis (PSC) is characterized by biliary damage and fibrosis. Multidrug resistance-2 gene knockout (Mdr2-/-) mice and PSC patients have increased histamine (HA) levels (synthesized by l-histidine decarboxylase, HDC) and HA receptor (HR) expression. Cholestatic HDC-/- mice display ameliorated biliary damage and hepatic fibrosis. The current study evaluated the effects of knockout of HDC-/- in Mdr2-/- mice (DKO) on biliary damage and hepatic fibrosis. WT, Mdr2-/- mice, and homozygous DKO mice were used. Selected DKO mice were treated with HA. We evaluated liver damage along with HDC expression and HA serum levels. Changes in ductular reaction were evaluated along with liver fibrosis, inflammation and bile acid signaling pathways. The expression of H1HR/PKC-α/TGF-β1 and H2HR/pERK/VEGF-C was determined. In vitro, cholangiocyte lines were treated with HA with/without H1/H2 inhibitors before measuring: H1/H2HR, TGF-β1, and VEGF-C expression. Knockout of HDC ameliorates hepatic damage, ductular reaction, fibrosis, inflammation, bile acid signaling and H1HR/PKC-α/TGF-β1 and H2HR/pERK/VEGF-C signaling. Reactivation of the HDC/HA axis increased these parameters. In vitro, stimulation with HA increased HR expression and PKC-α, TGF-β1, and VEGF-C expression, which was reduced with HR inhibitors. Our data demonstrate the key role for the HDC/HA axis in the management of PSC progression.
Collapse
|
17
|
Huang D, Liu H, Zhu A, Zhou Y, Li Y. Forebrain excitatory neuron-specific SENP2 knockout mouse displays hyperactivity, impaired learning and memory, and anxiolytic-like behavior. Mol Brain 2020; 13:59. [PMID: 32290845 PMCID: PMC7155287 DOI: 10.1186/s13041-020-00591-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Sentrin/SUMO-specific protease 2 (SENP2) is a member of SENPs family involved in maturation of SUMO precursors and deSUMOylation of specific target, and is highly expressed in the central nervous system (CNS). Although SENP2 has been shown to modulate embryonic development, fatty acid metabolism, atherosclerosis and epilepsy, the function of SENP2 in the CNS remains poorly understood. To address the role of SENP2 in the CNS and its potential involvement in neuropathology, we generated SENP2 conditional knockout mice by crossing floxed SENP2 mice with CaMKIIα-Cre transgenic mice. Behavioral tests revealed that SENP2 ablation induced hyper-locomotor activity, anxiolytic-like behaviors, spatial working memory impairment and fear-associated learning defect. In line with these observations, our RNA sequencing (RNA-seq) data identified a variety of differential expression genes that are particularly enriched in locomotion, learning and memory related biologic process. Taken together, our results indicated that SENP2 plays a critical role in emotional and cognitive regulation. This SENP2 conditional knockout mice model may help reveal novel mechanisms that underlie a variety of neuropsychiatric disorders associated with anxiety and cognition.
Collapse
Affiliation(s)
- Dehua Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Huiqing Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Aoxue Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yi Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
18
|
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
|
19
|
Annamalai B, Ragu Varman D, Horton RE, Daws LC, Jayanthi LD, Ramamoorthy S. Histamine Receptors Regulate the Activity, Surface Expression, and Phosphorylation of Serotonin Transporters. ACS Chem Neurosci 2020; 11:466-476. [PMID: 31916747 DOI: 10.1021/acschemneuro.9b00664] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Reuptake and clearance of released serotonin (5-HT) are critical in serotonergic neurotransmission. Serotonin transporter (SERT) is mainly responsible for clearing the extracellular 5-HT. Controlled trafficking, phosphorylation, and protein stability have been attributed to robust SERT activity. H3 histamine receptors (H3Rs) act in conjunction and regulate 5-HT release. H3Rs are expressed in the nervous system and located at the serotonergic terminals, where they act as heteroreceptors. Although histaminergic and serotonergic neurotransmissions are thought to be two separate events, whether H3Rs influence SERT in the CNS to control 5-HT reuptake has never been addressed. With a priori knowledge gained from our studies, we explored the possibility of using rat hippocampal synaptosomal preparations. We found that treatment with H3R/H4R-agonists immepip and (R)-(-)-α-methyl-histamine indeed resulted in a time- and concentration-dependent decrease in 5-HT transport. On the other hand, treatment with H3R/H4R-inverse agonist thioperamide caused a moderate increase in 5-HT uptake while blocking the inhibitory effect of H3R/H4R agonists. When investigated further, immepip treatment reduced the level of SERT on the plasma membrane and its phosphorylation. Likewise, CaMKII inhibitor KN93 or calcineurin inhibitor cyclosporine A also inhibited SERT function; however, an additive effect with immepip was not seen. High-speed in vivo chronoamperometry demonstrated that immepip delayed 5-HT clearance while thioperamide accelerated 5-HT clearance from the extracellular space. Immepip selectively inhibited SERT activity in the hippocampus and cortex but not in the striatum, midbrain, and brain stem. Thus, we report here a novel mechanism of regulating SERT activity by H3R-mediated CaMKII/calcineurin pathway in a brain-region-specific manner and perhaps synaptic 5-HT in the CNS that controls 5-HT clearance.
Collapse
Affiliation(s)
- Balasubramaniam Annamalai
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Rebecca E. Horton
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
20
|
Depienne C, Ciura S, Trouillard O, Bouteiller D, Leitão E, Nava C, Keren B, Marie Y, Guegan J, Forlani S, Brice A, Anheim M, Agid Y, Krack P, Damier P, Viallet F, Houeto JL, Durif F, Vidailhet M, Worbe Y, Roze E, Kabashi E, Hartmann A. Association of Rare Genetic Variants in Opioid Receptors with Tourette Syndrome. TREMOR AND OTHER HYPERKINETIC MOVEMENTS (NEW YORK, N.Y.) 2019; 9:tre-09-693. [PMID: 31824749 PMCID: PMC6878848 DOI: 10.7916/tohm.v0.693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/15/2019] [Indexed: 01/07/2023]
Abstract
Background Genes involved in Tourette syndrome (TS) remain largely unknown. We aimed to identify genetic factors contributing to TS in a French cohort of 120 individuals using a combination of hypothesis-driven and exome-sequencing approaches. Methods We first sequenced exons of SLITRK1-6 and HDC in the TS cohort and subsequently sequenced the exome of 12 individuals harboring rare variants in these genes to find additional rare variants contributing to the disorder under the hypothesis of oligogenic inheritance. We further screened three candidate genes (OPRK1, PCDH10, and NTSR2) preferentially expressed in the basal ganglia, and three additional genes involved in neurotensin and opioid signaling (OPRM1, NTS, and NTSR1), and compared variant frequencies in TS patients and 788 matched control individuals. We also investigated the impact of altering the expression of Oprk1 in zebrafish. Results Thirteen ultrarare missense variants of SLITRK1-6 and HDC were identified in 12 patients. Exome sequencing in these patients revealed rare possibly deleterious variants in 3,041 genes, 54 of which were preferentially expressed in the basal ganglia. Comparison of variant frequencies altering selected candidate genes in TS and control individuals revealed an excess of potentially disrupting variants in OPRK1, encoding the opioid kappa receptor, in TS patients. Accordingly, we show that downregulation of the Oprk1 orthologue in zebrafish induces a hyperkinetic phenotype in early development. Discussion These results support a heterogeneous and complex genetic etiology of TS, possibly involving rare variants altering the opioid pathway in some individuals, which could represent a novel therapeutic target in this disorder.
Collapse
Affiliation(s)
- Christel Depienne
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, DE
| | - Sorana Ciura
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Oriane Trouillard
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Delphine Bouteiller
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Elsa Leitão
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, DE
| | - Caroline Nava
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département de Génétique, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Boris Keren
- Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département de Génétique, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Yannick Marie
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Justine Guegan
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Sylvie Forlani
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Alexis Brice
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Mathieu Anheim
- Service de neurologie, CHU de Strasbourg, Hôpital de Hautepierre, Avenue Molière, 67200 Strasbourg Strasbourg, FR
| | - Yves Agid
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Paul Krack
- Service de Neurologie, CHU de Grenoble, Avenue Maquis du Grésivaudan, 38700 La Tronche, FR.,Center for Movement Disorders, Inselspital, University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Philippe Damier
- Service de Neurologie, CHU de Nantes, 5 Allée de l'Île Gloriette, 44093 Nantes, FR
| | - François Viallet
- Service de Neurologie, CRHU d'Aix-en-Provence, Avenue des Tamaris, 13100 Aix-en-Provence, FR
| | - Jean-Luc Houeto
- Service de Neurologie, CHU de Poitiers, 2 Rue de la Milétrie, 86021 Poitiers, FR
| | - Franck Durif
- Service de Neurologie, CHU de Clermont-Ferrand, CHU de Clermont-Ferrand, Hôpital Gabriel Montpied, 58 rue Montalembert, 63003 Clermont-Ferrand, FR
| | - Marie Vidailhet
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Yulia Worbe
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Emmanuel Roze
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Edor Kabashi
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| | - Andreas Hartmann
- INSERM, U 1127, CNRS UMR 7225, Faculté de Médecine de Sorbonne Université, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,Assistance Publique Hôpitaux de Paris (APHP), Hôpital Pitié-Salpêtrière, Département de Neurologie, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR.,AP-HP, Centre de Référence National Maladie Rare 'Syndrome Gilles de la Tourette', Hôpital Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75013 Paris, FR
| |
Collapse
|
21
|
Pittenger C, Pushkarskaya H, Gruner P. Animal models of OCD-relevant processes: an RDoC perspective. J Obsessive Compuls Relat Disord 2019; 23:100433. [PMID: 32322462 PMCID: PMC7176322 DOI: 10.1016/j.jocrd.2019.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Animal models have been invaluable tools in deciphering pathophysiology in many branches of medicine. Their application in the study of complex neuropsychiatric conditions such as obsessive-compulsive disorder (OCD), however, raises vexing interpretative challenges. The Research Domain Criteria (RDoC) approach of identifying dimensions of function and dysfunction that cut across syndromic diagnoses provides one potential path forward. We review some of the domains in the current RDoC matrix that may inform our understanding of patients with obsessions and compulsions, and how work in animal model systems is helping us to understand them. We focus on three specific RDoC constructs that may be particularly informative for our understanding of OCD: potential threat, habit, and cognitive control. In each case we review selected recent studies in animal models and their potential contribution to our understanding of OCD, and suggest directions for future research, informed by the animal studies. Such mechanistic work in animal models, in parallel with clinical studies refining our understanding of the relationship between these dimensional constructs and the symptomatology of particular groups of patients, may over time help us to generate a more comprehensive understanding of the pathogenesis and complexity of obsessions and compulsions.
Collapse
Affiliation(s)
- Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine
- Child Study Center, Yale University School of Medicine
- Interdepartmental Neuroscience Program, Yale University School of Medicine
| | | | - Patricia Gruner
- Department of Psychiatry, Yale University School of Medicine
| |
Collapse
|
22
|
Pittenger C. The histidine decarboxylase model of tic pathophysiology: a new focus on the histamine H 3 receptor. Br J Pharmacol 2019; 177:570-579. [PMID: 30714121 DOI: 10.1111/bph.14606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
Histamine dysregulation was implicated as a rare cause of Tourette syndrome and other tic disorders a decade ago by a landmark genetic study in a high density family pedigree, which implicated a hypomorphic mutation in the histidine decarboxylase (Hdc) gene as a rare but high penetrance genetic cause. Studies in Hdc knockout (KO) mice have confirmed that this mutation causes tic-relevant behavioural and neurochemical abnormalities that parallel what is seen in patients and thus validate the KO as a potentially informative model of tic pathophysiology. Recent studies have focused on the potential role of the histamine H3 receptor in this model, and by association in tic disorders and related neuropsychiatric conditions. The H3 receptor is up-regulated in the striatum in Hdc KO mice. As the H3 receptor has constitutive activity in the absence of ligand, this receptor up-regulation may have significant cellular effects despite the absence of neurotransmitter histamine in these mice. Activation in vivo of H3 receptors in wild type mice regulates signalling in striatal medium spiny neurons (MSNs) that interacts non-linearly with dopamine receptor signalling. Baseline signalling alterations in MSNs in Hdc KO mice resemble those seen after H3 receptor agonist treatment in wild type animals. H3 receptor agonist treatment in the KOs further accentuates most of these signalling abnormalities and produces behavioural stereotypy. Together, these data suggest the intriguing hypothesis that constitutive signalling by up-regulated H3 receptors explains many of the molecular and behavioural abnormalities seen in these animals. LINKED ARTICLES: This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.
Collapse
|
23
|
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: 2.2] [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
|
24
|
Rapanelli M, Frick L, Jindachomthong K, Xu J, Ohtsu H, Nairn AC, Pittenger C. Striatal Signaling Regulated by the H3R Histamine Receptor in a Mouse Model of tic Pathophysiology. Neuroscience 2018; 392:172-179. [PMID: 30278251 PMCID: PMC6204318 DOI: 10.1016/j.neuroscience.2018.09.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022]
Abstract
Histamine dysregulation has been identified as a rare genetic cause of tic disorders; mice with a knockout of the histidine decarboxylase (Hdc) gene represent a promising model of this pathophysiology. How alterations in the histamine system lead to neuropsychiatric disease, however, remains unclear. The H3R histamine receptor is elevated in the striatum of Hdc KO mice, and H3R agonists, acting in the dorsal striatum, trigger tic-like movements in the model. In wild-type mice, H3R in the dorsal striatum differentially regulates mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) signaling in D1R dopamine receptor-expressing striatonigral medium spiny neurons (dMSNs) and D2R dopamine receptor-expressing striatopallidal MSNs (iMSNs), respectively. We examined the effects of H3R agonist treatment on MSN signaling in the Hdc-KO model. In dMSNs, MAPK signaling was elevated at baseline in the Hdc-KO model, resembling what is seen after H3R activation in WT animals. Similarly, in iMSNs, Akt phosphorylation was reduced at baseline in the KO model, resembling what is seen after H3R activation in WT animals. H3R activation in Hdc-KO mice further enhanced the baseline effect on Akt phosphorylation in iMSNs but attenuated the abnormality in MAPK signaling in dMSNs. These observations support the hypothesis that constitutive activity of upregulated H3R receptors in the Hdc-KO model mediates the observed alterations in baseline MSN signaling; but further activation of H3R, which produces tic-like repetitive movements in the model, has more complex effects.
Collapse
Affiliation(s)
| | - Luciana Frick
- Department of Psychiatry, Yale University, United States
| | | | - Jian Xu
- Department of Psychiatry, Yale University, United States; Child Study Center, Yale University, United States
| | - Hiroshi Ohtsu
- Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Angus C Nairn
- Department of Psychiatry, Yale University, United States; Interdepartental Neuroscience Program, Yale University, United States
| | - Christopher Pittenger
- Department of Psychiatry, Yale University, United States; Child Study Center, Yale University, United States; Interdepartental Neuroscience Program, Yale University, United States.
| |
Collapse
|
25
|
Rossignoli G, Grottesi A, Bisello G, Montioli R, Borri Voltattorni C, Paiardini A, Bertoldi M. Cysteine 180 Is a Redox Sensor Modulating the Activity of Human Pyridoxal 5'-Phosphate Histidine Decarboxylase. Biochemistry 2018; 57:6336-6348. [PMID: 30346159 DOI: 10.1021/acs.biochem.8b00625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Histidine decarboxylase is a pyridoxal 5'-phosphate enzyme catalyzing the conversion of histidine to histamine, a bioactive molecule exerting its role in many modulatory processes. The human enzyme is involved in many physiological functions, such as neurotransmission, gastrointestinal track function, cell growth, and differentiation. Here, we studied the functional properties of the human enzyme and, in particular, the effects exerted at the protein level by two cysteine residues: Cys-180 and Cys-418. Surprisingly, the enzyme exists in an equilibrium between a reduced and an oxidized form whose extent depends on the redox state of Cys-180. Moreover, we determined that (i) the two enzymatic redox species exhibit modest structural changes in the coenzyme microenvironment and (ii) the oxidized form is slightly more active and stable than the reduced one. These data are consistent with the model proposed by bioinformatics analyses and molecular dynamics simulations in which the Cys-180 redox state could be responsible for a structural transition affecting the C-terminal domain reorientation leading to active site alterations. Furthermore, the biochemical properties of the purified C180S and C418S variants reveal that C180S behaves like the reduced form of the wild-type enzyme, while C418S is sensitive to reductants like the wild-type enzyme, thus allowing the identification of Cys-180 as the redox sensitive switch. On the other hand, Cys-418 appears to be a residue involved in aggregation propensity. A possible role for Cys-180 as a regulatory switch in response to different cellular redox conditions could be suggested.
Collapse
Affiliation(s)
- Giada Rossignoli
- Department of Neuroscience, Biomedicine and Movement , University of Verona , Strada Le Grazie, 8 , 37134 Verona , Italy
| | | | - Giovanni Bisello
- Department of Neuroscience, Biomedicine and Movement , University of Verona , Strada Le Grazie, 8 , 37134 Verona , Italy
| | - Riccardo Montioli
- Department of Neuroscience, Biomedicine and Movement , University of Verona , Strada Le Grazie, 8 , 37134 Verona , Italy
| | - Carla Borri Voltattorni
- Department of Neuroscience, Biomedicine and Movement , University of Verona , Strada Le Grazie, 8 , 37134 Verona , Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences "A. Rossi Fanelli" , University "La Sapienza", Rome , P.zale A. Moro 5 , 00185 Roma , Italy
| | - Mariarita Bertoldi
- Department of Neuroscience, Biomedicine and Movement , University of Verona , Strada Le Grazie, 8 , 37134 Verona , Italy
| |
Collapse
|
26
|
Adams TG, Kelmendi B, Brake CA, Gruner P, Badour CL, Pittenger C. The role of stress in the pathogenesis and maintenance of obsessive-compulsive disorder. ACTA ACUST UNITED AC 2018. [PMID: 29527593 PMCID: PMC5841259 DOI: 10.1177/2470547018758043] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Individuals with obsessive-compulsive disorder often identify psychosocial stress
as a factor that exacerbates their symptoms, and many trace the onset of
symptoms to a stressful period of life or a discrete traumatic incident.
However, the pathophysiological relationship between stress and
obsessive-compulsive disorder remains poorly characterized: it is unclear
whether trauma or stress is an independent cause of obsessive-compulsive
disorder symptoms, a triggering factor that interacts with a preexisting
diathesis, or simply a nonspecific factor that can exacerbate
obsessive-compulsive disorder along with other aspects of psychiatric
symptomatology. Nonetheless, preclinical research has demonstrated that stress
has conspicuous effects on corticostriatal and limbic circuitry. Specifically,
stress can lead to neuronal atrophy in frontal cortices (particularly the medial
prefrontal cortex), the dorsomedial striatum (caudate), and the hippocampus.
Stress can also result in neuronal hypertrophy in the dorsolateral striatum
(putamen) and amygdala. These neurobiological effects mirror reported neural
abnormalities in obsessive-compulsive disorder and may contribute to an
imbalance between goal-directed and habitual behavior, an imbalance that is
implicated in the pathogenesis and expression of obsessive-compulsive disorder
symptomatology. The modulation of corticostriatal and limbic circuits by stress
and the resultant imbalance between habit and goal-directed learning and
behavior offers a framework for investigating how stress may exacerbate or
trigger obsessive-compulsive disorder symptomatology.
Collapse
Affiliation(s)
- T G Adams
- School of Medicine - Department of Psychiatry, Yale University.,Clinical Neuroscience Division of the VA National Center for PTSD
| | - B Kelmendi
- School of Medicine - Department of Psychiatry, Yale University.,Clinical Neuroscience Division of the VA National Center for PTSD
| | - C A Brake
- University of Kentucky, Department of Psychology
| | - P Gruner
- School of Medicine - Department of Psychiatry, Yale University
| | - C L Badour
- University of Kentucky, Department of Psychology
| | - C Pittenger
- School of Medicine - Department of Psychiatry, Yale University.,Clinical Neuroscience Division of the VA National Center for PTSD.,Child Study Center, Yale University.,Department of Psychology, Yale University
| |
Collapse
|
27
|
Fernandez TV, State MW, Pittenger C. Tourette disorder and other tic disorders. HANDBOOK OF CLINICAL NEUROLOGY 2018; 147:343-354. [PMID: 29325623 DOI: 10.1016/b978-0-444-63233-3.00023-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tourette disorder is a developmental neuropsychiatric condition characterized by vocal and motor tics that can range in severity from mild to disabling. It represents one end of a spectrum of tic disorders and is estimated to affect 0.5-0.7% of the population. Accumulated evidence supports a substantial genetic contribution to disease risk, but the identification of genetic variants that confer risk has been challenging. Positive findings in candidate gene association studies have not replicated, and genomewide association studies have not generated signals of genomewide significance, in large part because of inadequate sample sizes. Rare mutations in several genes have been identified, but their causality is difficult to establish. As in other complex neuropsychiatric disorders, it is likely that Tourette disorder risk involves a combination of common, low-effect and rare, larger-effect variants in multiple genes acting together with environmental factors. With the ongoing collection of larger patient cohorts and the emergence of affordable high-throughput genomewide sequencing, progress is expected to accelerate in coming years.
Collapse
Affiliation(s)
- Thomas V Fernandez
- Child Study Center, Yale School of Medicine, New Haven, CT, United States
| | - Matthew W State
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, United States
| | - Christopher Pittenger
- Child Study Center, Yale School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University, New Haven, CT, United States.
| |
Collapse
|
28
|
Bortolato M, Pittenger C. Modeling tics in rodents: Conceptual challenges and paths forward. J Neurosci Methods 2017; 292:12-19. [PMID: 28237575 PMCID: PMC5568514 DOI: 10.1016/j.jneumeth.2017.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Recent advances in our understanding of the neurobiology of tics have led to the development of novel rodent models capturing different pathophysiological and phenotypic aspects of Tourette syndrome. The proliferation of these models, however, raises vexing questions on what standards should be adopted to assess their theoretical validity and empirical utility. Assessing the homology of a rodent motoric burst with a tic remains problematic, due to our incomplete knowledge of the underpinnings of tics, their high phenotypic complexity and variability, limitations in our ability test key aspects of tic phenomenology (such as premonitory sensory phenomena) in animals, and between-species differences in neuroanatomy and behavioral repertoire. These limitations underscore that any interpretation of behavioral output in an animal model cannot exclusively rely on the recognition of features that bear superficial resemblance with tics, but must be supported by other etiological and convergent phenomenological criteria. NEW METHOD Here, we discuss two complementary approaches for the study and validation of tic-like manifestations in rodents, based respectively on the use of contextual modulators and accompanying features of repetitive motor manifestations and on the reproduction of pathogenic factors. RESULTS Neither strategy can by itself provide convincing evidence that a model informatively recapitulates tic pathophysiology. Their combination holds promise to enhance the rigorous evaluation and translational relevance of rodent models of tic disorders. CONCLUSIONS This systematic consideration of different approaches to the validation and study of animal models of tic pathophysiology provides a framework for future work in this area.
Collapse
Affiliation(s)
- Marco Bortolato
- Department of Pharmacology and Toxicology, Interdepartmental Neuroscience Program, University of Utah, 30 S 2000 E, Skaggs Hall, Room 3916, Salt Lake City, UT, 84112, USA.
| | - Christopher Pittenger
- Department of Psychiatry, Department of Psychology, Child Study Center, Interdepartmental Neuroscience Program, Yale University, 34 Park Street, W315, New Haven, CT, 06519, USA.
| |
Collapse
|
29
|
Histamine modulation of the basal ganglia circuitry in the development of pathological grooming. Proc Natl Acad Sci U S A 2017; 114:6599-6604. [PMID: 28584117 DOI: 10.1073/pnas.1704547114] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aberrant histaminergic function has been proposed as a cause of tic disorders. A rare mutation in the enzyme that produces histamine (HA), histidine decarboxylase (HDC), has been identified in patients with Tourette syndrome (TS). Hdc knockout mice exhibit repetitive behavioral pathology and neurochemical characteristics of TS, establishing them as a plausible model of tic pathophysiology. Where, when, and how HA deficiency produces these effects has remained unclear: whether the contribution of HA deficiency to pathogenesis is acute or developmental, and where in the brain the relevant consequences of HA deficiency occur. Here, we address these key pathophysiological questions, using anatomically and cellularly targeted manipulations in mice. We report that specific ablation or chemogenetic silencing of histaminergic neurons in the tuberomammillary nucleus (TMN) of the hypothalamus leads to markedly elevated grooming, a form of repetitive behavioral pathology, and to elevated markers of neuronal activity in both dorsal striatum and medial prefrontal cortex. Infusion of HA directly into the striatum reverses this behavioral pathology, confirming that acute HA deficiency mediates the effect. Bidirectional chemogenetic regulation reveals that dorsal striatum neurons activated after TMN silencing are both sufficient to produce repetitive behavioral pathology and necessary for the full expression of the effect. Chemogenetic activation of TMN-regulated medial prefrontal cortex neurons, in contrast, increases locomotion and not grooming. These data confirm the centrality of striatal regulation by neurotransmitter HA in the adult in the production of pathological grooming.
Collapse
|
30
|
Shan L, Bao AM, Swaab DF. Changes in Histidine Decarboxylase, Histamine N-Methyltransferase and Histamine Receptors in Neuropsychiatric Disorders. Handb Exp Pharmacol 2017; 241:259-276. [PMID: 28233178 DOI: 10.1007/164_2016_125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Compared to other monoamine neurotransmitters, information on the association between the histaminergic system and neuropsychiatric disorders is scarce, resulting in a lack of histamine-related treatment for these disorders. The current chapter tries to combine information obtained from genetic studies, neuroimaging, post-mortem human brain studies and cerebrospinal fluid measurements with data from recent clinical trials on histamine receptor agonists and antagonists, with a view to determining the possible role of the histaminergic system in neuropsychiatric disorders and to pave the way for novel histamine-related therapeutic strategies.
Collapse
Affiliation(s)
- Ling Shan
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam, 1105 BA, The Netherlands
| | - Ai-Min Bao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, University of Amsterdam, Meibergdreef 47, Amsterdam, 1105 BA, The Netherlands.
| |
Collapse
|