1
|
Kos J, Langiu M, Hellyer SD, Gregory KJ. Pharmacology, Signaling and Therapeutic Potential of Metabotropic Glutamate Receptor 5 Negative Allosteric Modulators. ACS Pharmacol Transl Sci 2024; 7:3671-3690. [PMID: 39698283 PMCID: PMC11651194 DOI: 10.1021/acsptsci.4c00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 12/20/2024]
Abstract
Metabotropic glutamate receptors are a family of eight class C G protein-coupled receptors regulating higher order brain functions including cognition and motion. Metabotropic glutamate receptors have thus been heavily investigated as potential drug targets for treating neurological disorders. Drug discovery efforts directed toward metabotropic glutamate receptor subtype 5 (mGlu5) have been particularly fruitful, with a wealth of drug candidates and pharmacological tools identified. mGlu5 negative allosteric modulators (NAMs) are promising novel therapeutics for developmental, neuropsychiatric and neurodegenerative disorders (e.g., Alzheimer's Disease, Huntington's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, autism spectrum disorders, substance use disorders, stroke, anxiety and depression) and show promise in ameliorating adverse effects induced by other medications (e.g., L-dopa induced dyskinesia in Parkinson's Disease). However, despite preclinical success, mGlu5 NAMs are yet to reach the market due to poor safety and efficacy profiles in clinical trials. Herein, we review the physiology and signal transduction of mGlu5. We provide a comprehensive critique of therapeutic options with respect to mGlu5 inhibitors, spanning from orthosteric antagonists to NAMs. Finally, we address the challenges associated with drug development and highlight future directions to guide rational drug discovery of safe and effective novel therapeutics.
Collapse
Affiliation(s)
- Jackson
A. Kos
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Shane D. Hellyer
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Karen J. Gregory
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
- ARC
Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
2
|
Yu J, Deng X, Lin X, Xie L, Guo S, Lin X, Lin D. DST regulates cisplatin resistance in colorectal cancer via PI3K/Akt pathway. J Pharm Pharmacol 2024:rgae104. [PMID: 39419785 DOI: 10.1093/jpp/rgae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Dystonin (DST), a potential tumor suppressor gene, plays a crucial role in regulating cancer cell proliferation and resistance to chemotherapy. However, DST's specific role in colorectal cancer (CRC) has not been thoroughly investigated, and this study aims to elucidate its molecular role in modulating cisplatin (DDP) resistance in CRC. METHODS DST expression was analyzed in CRC tumors, DDP-resistant CRC tissues, paracancer tissues, and normal tissues. Lentiviral overexpression and shRNA knockdown were conducted in advanced CRC and DDP-resistant cell lines to assess cell viability, apoptosis, invasion, migration, proliferation, and angiogenesis. Xenograft mouse models studied DST's impact on CRC tumor growth and DDP resistance in vivo. RESULTS DST expression was significantly reduced in CRC tumor and DDP-resistant CRC tissues compared to paracancer and normal tissues (P < .001). Upregulating DST inhibited CRC and DDP-resistant cell viability, proliferation, invasion, and migration while promoting apoptosis. DST overexpression also reduced angiogenesis and attenuated DDP-induced cytotoxicity in CRC cells. Mechanistically, DST upregulation suppressed DDP resistance in CRC cells via the PI3K/Akt signaling pathway. DST upregulation reduced CRC tumor growth and mitigated DDP resistance, in vivo. CONCLUSION DST plays a crucial role in limiting CRC progression and overcoming DDP resistance, suggesting potential for targeted CRC therapies.
Collapse
Affiliation(s)
- Jianwei Yu
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xueqiong Deng
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xueqin Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Li Xie
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Sisi Guo
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Xiaoliang Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Dong Lin
- Department of Gastroenterology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan 364000, Fujian Province, China
| |
Collapse
|
3
|
Kilic-Berkmen G, Kim H, Chen D, Yeo CI, Dinasarapu AR, Scorr LM, Yeo WH, Peterson DA, Williams H, Ruby A, Mills R, Jinnah HA. An Exploratory, Randomized, Double-Blind Clinical Trial of Dipraglurant for Blepharospasm. Mov Disord 2024; 39:738-745. [PMID: 38310362 PMCID: PMC11045316 DOI: 10.1002/mds.29734] [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: 09/25/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Blepharospasm is treated with botulinum toxin, but obtaining satisfactory results is sometimes challenging. OBJECTIVE The aim is to conduct an exploratory trial of oral dipraglurant for blepharospasm. METHODS This study was an exploratory, phase 2a, randomized, double-blind, placebo-controlled trial of 15 participants who were assigned to receive a placebo or dipraglurant (50 or 100 mg) and assessed over 2 days, 1 and 2 hours following dosing. Outcome measures included multiple scales rated by clinicians or participants, digital video, and a wearable sensor. RESULTS Dipraglurant was well tolerated, with no obvious impact on any of the measurement outcomes. Power analyses suggested fewer subjects would be required for studies using a within-subject versus independent group design, especially for certain measures. Some outcome measures appeared more suitable than others. CONCLUSION Although dipraglurant appeared well tolerated, it did not produce a trend for clinical benefit. The results provide valuable information for planning further trials in blepharospasm. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Gamze Kilic-Berkmen
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hodam Kim
- IEN Center for Human-Centric Interfaces and Engineering at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Dongdong Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Cameron I. Yeo
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ashok R. Dinasarapu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Laura M. Scorr
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Woon-Hong Yeo
- IEN Center for Human-Centric Interfaces and Engineering at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Institute for Materials, Neural Engineering Center, Institute for Robotics and Intelligent Machines, Georgia Institute of Technology, Atlanta, GA, USA
| | - David A. Peterson
- Institute for Neural Computation, University of California in San Diego, La Jolla, CA, United States
| | - Hilde Williams
- Drug Development Consultant, Addex Pharmaceuticals Inc. Geneva Switzerland
| | - April Ruby
- Drug Development Consultant, Addex Pharmaceuticals Inc. Geneva Switzerland
| | - Roger Mills
- Drug Development Consultant, Addex Pharmaceuticals Inc. Geneva Switzerland
| | - H. A. Jinnah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
El Atiallah I, Ponterio G, Meringolo M, Martella G, Sciamanna G, Tassone A, Montanari M, Mancini M, Castagno AN, Yu-Taeger L, Nguyen HHP, Bonsi P, Pisani A. Loss-of-function of GNAL dystonia gene impairs striatal dopamine receptors-mediated adenylyl cyclase/ cyclic AMP signaling pathway. Neurobiol Dis 2024; 191:106403. [PMID: 38182074 DOI: 10.1016/j.nbd.2024.106403] [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: 11/21/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Loss-of-function mutations in the GNAL gene are responsible for DYT-GNAL dystonia. However, how GNAL mutations contribute to synaptic dysfunction is still unclear. The GNAL gene encodes the Gαolf protein, an isoform of stimulatory Gαs enriched in the striatum, with a key role in the regulation of cAMP signaling. Here, we used a combined biochemical and electrophysiological approach to study GPCR-mediated AC-cAMP cascade in the striatum of the heterozygous GNAL (GNAL+/-) rat model. We first analyzed adenosine type 2 (A2AR), and dopamine type 1 (D1R) receptors, which are directly coupled to Gαolf, and observed that the total levels of A2AR were increased, whereas D1R level was unaltered in GNAL+/- rats. In addition, the striatal isoform of adenylyl cyclase (AC5) was reduced, despite unaltered basal cAMP levels. Notably, the protein expression level of dopamine type 2 receptor (D2R), that inhibits the AC5-cAMP signaling pathway, was also reduced, similar to what observed in different DYT-TOR1A dystonia models. Accordingly, in the GNAL+/- rat striatum we found altered levels of the D2R regulatory proteins, RGS9-2, spinophilin, Gβ5 and β-arrestin2, suggesting a downregulation of D2R signaling cascade. Additionally, by analyzing the responses of striatal cholinergic interneurons to D2R activation, we found that the receptor-mediated inhibitory effect is significantly attenuated in GNAL+/- interneurons. Altogether, our findings demonstrate a profound alteration in the A2AR/D2R-AC-cAMP cascade in the striatum of the rat DYT-GNAL dystonia model, and provide a plausible explanation for our previous findings on the loss of dopamine D2R-dependent corticostriatal long-term depression.
Collapse
Affiliation(s)
- Ilham El Atiallah
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Mancini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Fondazione Mondino, Pavia, Italy
| | - Antonio N Castagno
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Fondazione Mondino, Pavia, Italy
| | - Libo Yu-Taeger
- Department of Human Genetics, Ruhr University Bochum, Germany
| | | | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Fondazione Mondino, Pavia, Italy.
| |
Collapse
|
5
|
Fan Y, Si Z, Wang L, Zhang L. DYT- TOR1A dystonia: an update on pathogenesis and treatment. Front Neurosci 2023; 17:1216929. [PMID: 37638318 PMCID: PMC10448058 DOI: 10.3389/fnins.2023.1216929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
DYT-TOR1A dystonia is a neurological disorder characterized by involuntary muscle contractions and abnormal movements. It is a severe genetic form of dystonia caused by mutations in the TOR1A gene. TorsinA is a member of the AAA + family of adenosine triphosphatases (ATPases) involved in a variety of cellular functions, including protein folding, lipid metabolism, cytoskeletal organization, and nucleocytoskeletal coupling. Almost all patients with TOR1A-related dystonia harbor the same mutation, an in-frame GAG deletion (ΔGAG) in the last of its 5 exons. This recurrent variant results in the deletion of one of two tandem glutamic acid residues (i.e., E302/303) in a protein named torsinA [torsinA(△E)]. Although the mutation is hereditary, not all carriers will develop DYT-TOR1A dystonia, indicating the involvement of other factors in the disease process. The current understanding of the pathophysiology of DYT-TOR1A dystonia involves multiple factors, including abnormal protein folding, signaling between neurons and glial cells, and dysfunction of the protein quality control system. As there are currently no curative treatments for DYT-TOR1A dystonia, progress in research provides insight into its pathogenesis, leading to potential therapeutic and preventative strategies. This review summarizes the latest research advances in the pathogenesis, diagnosis, and treatment of DYT-TOR1A dystonia.
Collapse
Affiliation(s)
- Yuhang Fan
- Department of Neurology, the Second Hospital of Jilin University, Changchun, China
| | - Zhibo Si
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Linlin Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lei Zhang
- Department of Neurology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Steel D, Reid KM, Pisani A, Hess EJ, Fox S, Kurian MA. Advances in targeting neurotransmitter systems in dystonia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:217-258. [PMID: 37482394 DOI: 10.1016/bs.irn.2023.06.002] [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: 07/25/2023]
Abstract
Dystonia is characterised as uncontrolled, often painful involuntary muscle contractions that cause abnormal postures and repetitive or twisting movements. These movements can be continuous or sporadic and affect different parts of the body and range in severity. Dystonia and its related conditions present a huge cause of neurological morbidity worldwide. Although therapies are available, achieving optimal symptom control without major unwanted effects remains a challenge. Most pharmacological treatments for dystonia aim to modulate the effects of one or more neurotransmitters in the central nervous system, but doing so effectively and with precision is far from straightforward. In this chapter we discuss the physiology of key neurotransmitters, including dopamine, noradrenaline, serotonin (5-hydroxytryptamine), acetylcholine, GABA, glutamate, adenosine and cannabinoids, and their role in dystonia. We explore the ways in which existing pharmaceuticals as well as novel agents, currently in clinical trial or preclinical development, target dystonia, and their respective advantages and disadvantages. Finally, we discuss current and emerging genetic therapies which may be used to treat genetic forms of dystonia.
Collapse
Affiliation(s)
- Dora Steel
- UCL GOS Institute of Child Health (Zayed Centre for Research into Rare Diseases in Children), London, United Kingdom; Great Ormond Street Hospital for Children, London, United Kingdom
| | - Kimberley M Reid
- UCL GOS Institute of Child Health (Zayed Centre for Research into Rare Diseases in Children), London, United Kingdom
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Mondino Foundation, Pavia, Italy
| | - Ellen J Hess
- Emory University School of Medicine, CA, United States
| | - Susan Fox
- Movement Disorders Clinic, Toronto Western Hospital, University of Toronto, ON, Canada
| | - Manju A Kurian
- UCL GOS Institute of Child Health (Zayed Centre for Research into Rare Diseases in Children), London, United Kingdom; Great Ormond Street Hospital for Children, London, United Kingdom.
| |
Collapse
|
7
|
El Atiallah I, Bonsi P, Tassone A, Martella G, Biella G, Castagno AN, Pisani A, Ponterio G. Synaptic Dysfunction in Dystonia: Update From Experimental Models. Curr Neuropharmacol 2023; 21:2310-2322. [PMID: 37464831 PMCID: PMC10556390 DOI: 10.2174/1570159x21666230718100156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 07/20/2023] Open
Abstract
Dystonia, the third most common movement disorder, refers to a heterogeneous group of neurological diseases characterized by involuntary, sustained or intermittent muscle contractions resulting in repetitive twisting movements and abnormal postures. In the last few years, several studies on animal models helped expand our knowledge of the molecular mechanisms underlying dystonia. These findings have reinforced the notion that the synaptic alterations found mainly in the basal ganglia and cerebellum, including the abnormal neurotransmitters signalling, receptor trafficking and synaptic plasticity, are a common hallmark of different forms of dystonia. In this review, we focus on the major contribution provided by rodent models of DYT-TOR1A, DYT-THAP1, DYT-GNAL, DYT/ PARK-GCH1, DYT/PARK-TH and DYT-SGCE dystonia, which reveal that an abnormal motor network and synaptic dysfunction represent key elements in the pathophysiology of dystonia.
Collapse
Affiliation(s)
- Ilham El Atiallah
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Antonio N. Castagno
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
8
|
Abu-hadid O, Jimenez-Shahed J. An overview of the pharmacotherapeutics for dystonia: advances over the past decade. Expert Opin Pharmacother 2022; 23:1927-1940. [DOI: 10.1080/14656566.2022.2147823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- O. Abu-hadid
- Icahn School of Medicine at Mount Sinai, New York City, NY
| | | |
Collapse
|
9
|
Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav 2022; 219:173446. [PMID: 35987339 DOI: 10.1016/j.pbb.2022.173446] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacological modulation of glutamate has long been considered to be of immense therapeutic utility. The metabotropic glutamate receptors (mGluRs) are potential targets for safely altering glutamate-driven excitation. Data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders. For each of the three mGluR groups, compounds have been constructed that produce either potentiation or functional blockade. PET ligands for mGlu5Rs have been studied in a range of patient populations and several mGlu5R antagonists have been tested for potential efficacy in patients including mavoglurant, diploglurant, basimglurant, GET 73, and ADX10059. Efficacy with mGlu5R antagonists has been reported in trials with patients with gastroesophageal reflux disease; data from patients with Parkinson's disease or Fragile X syndrome have not been as robust as hoped. Fenobam was approved for use as an anxiolytic prior to its recognition as an mGlu5R antagonist. mGlu2/3R agonists (pomaglumated methionil) and mGlu2R agonists (JNJ-40411813, AZD 8529, and LY2979165) have been studied in patients with schizophrenia with promising but mixed results. Antagonists of mGlu2/3Rs (decoglurant and TS-161) have been studied in depression where TS-161 has advanced into a planned Phase 2 study in treatment-resistant depression. The Group III mGluRs are the least developed of the mGluR receptor targets. The mGlu4R potentiator, foliglurax, did not meet its primary endpoint in patients with Parkinson's disease. Ongoing efforts to develop mGluR-targeted compounds continue to promise these glutamate modulators as medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | - Kamal P Pandey
- Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| |
Collapse
|
10
|
Scarduzio M, Hess EJ, Standaert DG, Eskow Jaunarajs KL. Striatal synaptic dysfunction in dystonia and levodopa-induced dyskinesia. Neurobiol Dis 2022; 166:105650. [DOI: 10.1016/j.nbd.2022.105650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
|
11
|
Sciamanna G, El Atiallah I, Montanari M, Pisani A. Plasticity, genetics and epigenetics in dystonia: An update. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:199-206. [PMID: 35034734 DOI: 10.1016/b978-0-12-819410-2.00011-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dystonia represents a group of movement disorders characterized by involuntary muscle contractions that result in abnormal posture and twisting movements. In the last 20 years several animal models have been generated, greatly improving our knowledge of the neural and molecular mechanism underlying this pathological condition, but the pathophysiology remains still poorly understood. In this review we will discuss recent genetic factors related to dystonia and the current understanding of synaptic plasticity alterations reported by both clinical and experimental research. We will also present recent evidence involving epigenetics mechanisms in dystonia.
Collapse
Affiliation(s)
- Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Ilham El Atiallah
- Department of Systems Medicine, University of Rome 2 Tor Vergata, Rome, Italy
| | - Martina Montanari
- Department of Systems Medicine, University of Rome 2 Tor Vergata, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Movement Disorders Research Center, IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
12
|
Cirnaru MD, Creus-Muncunill J, Nelson S, Lewis TB, Watson J, Ellerby LM, Gonzalez-Alegre P, Ehrlich ME. Striatal Cholinergic Dysregulation after Neonatal Decrease in X-Linked Dystonia Parkinsonism-Related TAF1 Isoforms. Mov Disord 2021; 36:2780-2794. [PMID: 34403156 DOI: 10.1002/mds.28750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND X-linked dystonia parkinsonism is a generalized, progressive dystonia followed by parkinsonism with onset in adulthood and accompanied by striatal neurodegeneration. Causative mutations are located in a noncoding region of the TATA-box binding protein-associated factor 1 (TAF1) gene and result in aberrant splicing. There are 2 major TAF1 isoforms that may be decreased in symptomatic patients, including the ubiquitously expressed canonical cTAF1 and the neuronal-specific nTAF1. OBJECTIVE The objective of this study was to determine the behavioral and transcriptomic effects of decreased cTAF1 and/or nTAF1 in vivo. METHODS We generated adeno-associated viral (AAV) vectors encoding microRNAs targeting Taf1 in a splice-isoform selective manner. We performed intracerebroventricular viral injections in newborn mice and rats and intrastriatal infusions in 3-week-old rats. The effects of Taf1 knockdown were assayed at 4 months of age with evaluation of motor function, histology, and RNA sequencing of the striatum, followed by its validation. RESULTS We report motor deficits in all cohorts, more pronounced in animals injected at P0, in which we also identified transcriptomic alterations in multiple neuronal pathways, including the cholinergic synapse. In both species, we show a reduced number of striatal cholinergic interneurons and their marker mRNAs after Taf1 knockdown in the newborn. CONCLUSION This study provides novel information regarding the requirement for TAF1 in the postnatal maintenance of striatal cholinergic neurons, the dysfunction of which is involved in other inherited forms of dystonia. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shareen Nelson
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Travis B Lewis
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jaime Watson
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, California, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
13
|
Rescue of striatal long-term depression by chronic mGlu5 receptor negative allosteric modulation in distinct dystonia models. Neuropharmacology 2021; 192:108608. [PMID: 33991565 DOI: 10.1016/j.neuropharm.2021.108608] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
An impairment of long-term synaptic plasticity is considered as a peculiar endophenotype of distinct forms of dystonia, a common, disabling movement disorder. Among the few therapeutic options, broad-spectrum antimuscarinic drugs are utilized, aimed at counteracting abnormal striatal acetylcholine-mediated transmission, which plays a crucial role in dystonia pathophysiology. We previously demonstrated a complete loss of long-term synaptic depression (LTD) at corticostriatal synapses in rodent models of two distinct forms of isolated dystonia, resulting from mutations in the TOR1A (DYT1), and GNAL (DYT25) genes. In addition to anticholinergic agents, the aberrant excitability of striatal cholinergic cells can be modulated by group I metabotropic glutamate receptor subtypes (mGlu1 and 5). Here, we tested the efficacy of the negative allosteric modulator (NAM) of metabotropic glutamate 5 (mGlu) receptor, dipraglurant (ADX48621) on striatal LTD. We show that, whereas acute treatment failed to rescue LTD, chronic dipraglurant rescued this form of synaptic plasticity both in DYT1 mice and GNAL rats. Our analysis of the pharmacokinetic profile of dipraglurant revealed a relatively short half-life, which led us to uncover a peculiar time-course of recovery based on the timing from last dipraglurant injection. Indeed, striatal spiny projection neurons (SPNs) recorded within 2 h from last administration showed full expression of synaptic plasticity, whilst the extent of recovery progressively diminished when SPNs were recorded 4-6 h after treatment. Our findings suggest that distinct dystonia genes may share common signaling pathway dysfunction. More importantly, they indicate that dipraglurant might be a potential novel therapeutic agent for this disabling disorder.
Collapse
|
14
|
Fox SH, Swan M, Jinnah HA, de Freitas MET, de Oliveira LM, Al-Shorafat D, Fernandez HH, Kompoliti K, Comella C. An Open-Label Phase 2a Study to Evaluate the Safety and Tolerability of Perampanel in Cervical Dystonia. Mov Disord Clin Pract 2021; 8:743-749. [PMID: 34307747 DOI: 10.1002/mdc3.13229] [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] [Received: 03/02/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 11/11/2022] Open
Abstract
Background Cervical dystonia (CD) is the most common focal isolated dystonia. Preclinical studies report that AMPA-selective glutamate receptor antagonists improve dystonia. Perampanel is a clinically available, AMPA receptor antagonist that has shown efficacy and safety in epilepsy. Objectives To determine safety and tolerability of perampanel in CD. Methods We performed a phase 2a, open-label, multicenter study to evaluate tolerability and safety of perampanel in CD. Included subjects had primary CD; those on botulinum toxin were 8 weeks post last injection. All subjects received perampanel 2 mg/day, titrated 2 mg weekly over 6 weeks, to maximum 12 mg/day; maintenance phase was 4 weeks, ending at week 10. Primary endpoints included tolerability, defined as ability to remain on perampanel for the maintenance period, at any dose level and safety, determined from adverse events (AEs) collected at each visit. Secondary exploratory endpoints included Toronto Western Spasmodic Torticollis Rating Scale (TWSTRS), quality of life (cervical dystonia impact profile [CIDP]-58) and Clinical Global Impression of change (CGI). Results CD participants (n = 25) were recruited. Eight subjects withdrew; 4 because of AEs, 3 for other reasons and 1 lost to follow up. One subject tolerated 12 mg/day. Eight subjects (30.8%) tolerated 2 mg, whereas 19.2% tolerated 4 mg/day, and 15.4% tolerated 6 mg or 8 mg/day. All subjects experienced AEs. The most common AEs were dizziness, imbalance, and irritability. Exploratory endpoints of TWSTRS showed some improved pain scores and CIDP-58 improved sleep. Conclusions Tolerability to perampanel was variable in CD subjects. Lower doses would be considered for future studies in this population.
Collapse
Affiliation(s)
- Susan H Fox
- The Edmond J Safra Program in Parkinson Disease, Toronto Western Hospital Toronto Ontario Canada.,Krembil Brain Institute, University Health Network Toronto Ontario Canada.,Division of Neurology University of Toronto Toronto Ontario Canada
| | - Matthew Swan
- Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai New York New York USA
| | - Hyder A Jinnah
- Department of Human Genetics and Pediatrics Emory University Atlanta Georgia USA
| | - Maria E T de Freitas
- The Edmond J Safra Program in Parkinson Disease, Toronto Western Hospital Toronto Ontario Canada.,Krembil Brain Institute, University Health Network Toronto Ontario Canada.,Division of Neurology University of Toronto Toronto Ontario Canada
| | - Lais M de Oliveira
- The Edmond J Safra Program in Parkinson Disease, Toronto Western Hospital Toronto Ontario Canada.,Krembil Brain Institute, University Health Network Toronto Ontario Canada.,Division of Neurology University of Toronto Toronto Ontario Canada
| | - Duha Al-Shorafat
- The Edmond J Safra Program in Parkinson Disease, Toronto Western Hospital Toronto Ontario Canada.,Krembil Brain Institute, University Health Network Toronto Ontario Canada.,Division of Neurology University of Toronto Toronto Ontario Canada
| | | | | | | |
Collapse
|
15
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
16
|
Perl S, Richter F, Richter A. Striatal and cortical metabotropic glutamate 5 receptor expression and behavioral effects of the positive allosteric modulator CDPPB in a model of DYT1 dystonia. Pharmacol Biochem Behav 2020; 196:172977. [PMID: 32615137 DOI: 10.1016/j.pbb.2020.172977] [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: 04/02/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022]
Abstract
The metabotropic glutamate 5 (mGlu5) receptor is critically involved in corticostriatal plasticity which is disturbed in various animal models of dystonia. Recently, the positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) exerted prodyskinetic effects in a phenotypic model of episodic dystonia. In the DYT1 knock-in (KI) mouse, a model for a persistent type of dystonia, previous ex vivo electrophysiological experiments indicated that mGlu5 receptors are involved in abnormal striatal plasticity. Therefore, in the present study we examined the mGlu5 receptor expression in the striatum and cortex of DYT1 KI mice in comparison with wildtype littermates. By immunohistochemistry (IHC) we found a lower expression of mGlu5 receptors in the cortex (16%) and ventral striatum (10%) but not in the whole striatum of DYT1 KI mice, while mRNA levels were merely lower in the striatum of DYT1 KI mice (43%). However, mGlu5 receptor protein levels measured by western blotting showed no significant differences in tissue of the whole striatum and in the cortex between both genotypes. Since DYT1 KI mice do not exhibit dystonic symptoms, we investigated if CDPPB provokes dystonia or dyskinesia. CDPPB (10, 20 and 30 mg/kg intraperitoneal, i.p.) did not induce abnormal movements and the locomotor activity did not differ between DYT1 KI and wildtype mice. The present data do not provide evidence for a crucial role of the mGlu5 receptor in the pathophysiology of DYT1 dystonia, but corticostriatal changes are in line with the hypothesis of maladaptive plasticity in dystonia.
Collapse
Affiliation(s)
- Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
17
|
Structure-based discovery and development of metabotropic glutamate receptor 5 negative allosteric modulators. ADVANCES IN PHARMACOLOGY 2020; 88:35-58. [PMID: 32416871 DOI: 10.1016/bs.apha.2020.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
The metabotropic glutamate (mGlu) receptors are a family of eight class C G protein-coupled receptors (GPCRs) which modulate cell signaling and synaptic transmission to the major excitatory neurotransmitter l-glutamate (l-glutamic acid). Due to their role in modulating glutamate response, their widespread distribution in the central nervous system (CNS) and some evidence of dysregulation in disease, the mGlu receptors have become attractive pharmacological targets. As the orthosteric (glutamate) binding site is highly conserved across the eight mGlu receptors, it is difficult not only to generate ligands with subtype selectivity but, due to the nature of the binding site, with suitable drug-like properties to allow oral bioavailability and CNS penetration. Selective pharmacological targeting of a single receptor subtype can be achieved by targeting alternative (allosteric) binding sites. The nature of the allosteric binding pockets allows ligands to be developed that have good physical chemical properties as evidenced by several allosteric modulators of mGlu receptors entering clinical trials. The first negative allosteric modulators of the metabotropic glutamate 5 (mGlu5) receptor were discovered from high throughput screening activities. An alternative approach to drug discovery is to use structural knowledge to enable structure-based drug design (SBDD), which allows the design of molecules in a more rational, rather than empirical, fashion. Here we will describe the process of SBDD in the discovery of the mGlu5 negative allosteric modulator HTL0014242 and describe how knowledge of receptor structure can also be used to gain insights into the receptor activation mechanisms.
Collapse
|
18
|
Imbriani P, Ponterio G, Tassone A, Sciamanna G, El Atiallah I, Bonsi P, Pisani A. Models of dystonia: an update. J Neurosci Methods 2020; 339:108728. [PMID: 32289333 DOI: 10.1016/j.jneumeth.2020.108728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
Although dystonia represents the third most common movement disorder, its pathophysiology remains still poorly understood. In the past two decades, multiple models have been generated, improving our knowledge on the molecular and cellular bases of this heterogeneous group of movement disorders. In this short survey, we will focus on recently generated novel models of DYT1 dystonia, the most common form of genetic, "isolated" dystonia. These models clearly indicate the existence of multiple signaling pathways affected by the protein mutation causative of DYT1 dystonia, torsinA, paving the way for potentially multiple, novel targets for pharmacological intervention.
Collapse
Affiliation(s)
- P Imbriani
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - G Ponterio
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - A Tassone
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - G Sciamanna
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - I El Atiallah
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - P Bonsi
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - A Pisani
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
19
|
Li J, Liang CC, Pappas SS, Dauer WT. TorsinB overexpression prevents abnormal twisting in DYT1 dystonia mouse models. eLife 2020; 9:e54285. [PMID: 32202496 PMCID: PMC7141835 DOI: 10.7554/elife.54285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic redundancy can be exploited to identify therapeutic targets for inherited disorders. We explored this possibility in DYT1 dystonia, a neurodevelopmental movement disorder caused by a loss-of-function (LOF) mutation in the TOR1A gene encoding torsinA. Prior work demonstrates that torsinA and its paralog torsinB have conserved functions at the nuclear envelope. This work established that low neuronal levels of torsinB dictate the neuronal selective phenotype of nuclear membrane budding. Here, we examined whether torsinB expression levels impact the onset or severity of abnormal movements or neuropathological features in DYT1 mouse models. We demonstrate that torsinB levels bidirectionally regulate these phenotypes. Reducing torsinB levels causes a dose-dependent worsening whereas torsinB overexpression rescues torsinA LOF-mediated abnormal movements and neurodegeneration. These findings identify torsinB as a potent modifier of torsinA LOF phenotypes and suggest that augmentation of torsinB expression may retard or prevent symptom development in DYT1 dystonia.
Collapse
Affiliation(s)
- Jay Li
- Medical Scientist Training Program, University of MichiganAnn ArborUnited States
- Cellular and Molecular Biology Graduate Program, University of MichiganAnn ArborUnited States
| | - Chun-Chi Liang
- Department of Neurology, University of MichiganAnn ArborUnited States
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| | - William T Dauer
- Department of Neurology, University of MichiganAnn ArborUnited States
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| |
Collapse
|
20
|
Gienger M, Hübner H, Löber S, König B, Gmeiner P. Structure-based development of caged dopamine D 2/D 3 receptor antagonists. Sci Rep 2020; 10:829. [PMID: 31965029 PMCID: PMC6972920 DOI: 10.1038/s41598-020-57770-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Dopamine is a neurotransmitter of great physiological relevance. Disorders in dopaminergic signal transduction are associated with psychiatric and neurological pathologies such as Parkinson’s disease, schizophrenia and substance abuse. Therefore, a detailed understanding of dopaminergic neurotransmission may provide access to novel therapeutic strategies for the treatment of these diseases. Caged compounds with photoremovable groups represent molecular tools to investigate a biological target with high spatiotemporal resolution. Based on the crystal structure of the D3 receptor in complex with eticlopride, we have developed caged D2/D3 receptor ligands by rational design. We initially found that eticlopride, a widely used D2/D3 receptor antagonist, was photolabile and therefore is not suitable for caging. Subtle structural modification of the pharmacophore led us to the photostable antagonist dechloroeticlopride, which was chemically transformed into caged ligands. Among those, the 2-nitrobenzyl derivative 4 (MG307) showed excellent photochemical stability, pharmacological behavior and decaging properties when interacting with dopamine receptor-expressing cells.
Collapse
Affiliation(s)
- Marie Gienger
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Stefan Löber
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Burkhard König
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany.
| |
Collapse
|
21
|
A Novel Transgenic Mouse Model to Investigate the Cell-Autonomous Effects of torsinA(ΔE) Expression in Striatal Output Neurons. Neuroscience 2019; 422:1-11. [PMID: 31669362 DOI: 10.1016/j.neuroscience.2019.09.007] [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: 05/24/2019] [Revised: 09/04/2019] [Accepted: 09/10/2019] [Indexed: 11/22/2022]
Abstract
Dystonia is a disabling neurological syndrome characterized by abnormal movements and postures that result from intermittent or sustained involuntary muscle contractions; mutations of DYT1/TOR1A are the most common cause of childhood-onset, generalized, inherited dystonia. Patient and mouse model data strongly support dysregulation of the nigrostriatal dopamine neurotransmission circuit in the presence of the DYT1-causing mutation. To determine striatal medium spiny neuron (MSN) cell-autonomous and non-cell autonomous effects relevant to dopamine transmission, we created a transgenic mouse in which expression of mutant torsinA in forebrain is restricted to MSNs. We assayed electrically evoked and cocaine-enhanced dopamine release and locomotor activity, dopamine uptake, gene expression of dopamine-associated neuropeptides and receptors, and response to the muscarinic cholinergic antagonist, trihexyphenidyl. We found that over-expression of mutant torsinA in MSNs produces complex cell-autonomous and non-cell autonomous alterations in nigrostriatal dopaminergic and intrastriatal cholinergic function, similar to that found in pan-cellular DYT1 mouse models. These data introduce targets for future studies to identify which are causative and which are compensatory in DYT1 dystonia, and thereby aid in defining appropriate therapies.
Collapse
|
22
|
Perl S, Richter F, Gericke B, Richter A. Expression of metabotropic glutamate 5 receptors in the striatum and cortex and effects of modulators on the severity of dystonia in the phenotypic dt sz model. Eur J Pharmacol 2019; 859:172527. [PMID: 31283933 DOI: 10.1016/j.ejphar.2019.172527] [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: 01/09/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
The metabotropic glutamate 5 (mGlu5) receptor has been suggested as therapeutic target for L-Dopa-induced dyskinesia which is often associated with dystonic symptoms. Therefore, we investigated the acute effects of the non-competitive mGlu5 receptor antagonist fenobam as well as the positive modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) on the severity of inherited dystonia in the mutant dtsz hamster, a phenotypic model with age-dependent episodes of dystonia. Fenobam did not exert significant antidystonic effects (20-50 mg/kg intraperinoneal, i.p.). CDPPB (10, 20 mg/kg i.p.) which was expected to worsen dystonia also failed to show any effects on the severity of dystonia. Interestingly, CDPPB caused axial dyskinesia in addition to the dystonic symptoms in mutant hamsters. This adverse effect could not be observed in non-dystonic control hamsters, indicating possible changes in the expression of mGlu5 receptors in dystonic hamsters. The mGlu5 receptor mRNA did not differ between the dtsz mutant and control hamsters, while immunohistochemical studies indicated that the mGlu5 receptor expression was about 35% higher in striatum and cortex of mutant hamsters at the age of high dystonia severity scores, notably not after spontaneous remission of dystonia, compared to age-matched controls. This difference in mGlu5 receptor protein may be due to altered protein conformation instead of protein level, as western blots revealed similar amounts of monomeric and dimeric protein in mutant hamsters versus control. Thus, the present data do not provide clear evidence for an important role of the mGlu5 receptor in the pathophysiology and as a therapeutic target for types of inherited dystonia.
Collapse
Affiliation(s)
- Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany; Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, D-30559, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, D-30559, Hannover, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
23
|
The neurobiological basis for novel experimental therapeutics in dystonia. Neurobiol Dis 2019; 130:104526. [PMID: 31279827 DOI: 10.1016/j.nbd.2019.104526] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/13/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022] Open
Abstract
Dystonia is a movement disorder characterized by involuntary muscle contractions, twisting movements, and abnormal postures that may affect one or multiple body regions. Dystonia is the third most common movement disorder after Parkinson's disease and essential tremor. Despite its relative frequency, small molecule therapeutics for dystonia are limited. Development of new therapeutics is further hampered by the heterogeneity of both clinical symptoms and etiologies in dystonia. Recent advances in both animal and cell-based models have helped clarify divergent etiologies in dystonia and have facilitated the identification of new therapeutic targets. Advances in medicinal chemistry have also made available novel compounds for testing in biochemical, physiological, and behavioral models of dystonia. Here, we briefly review motor circuit anatomy and the anatomical and functional abnormalities in dystonia. We then discuss recently identified therapeutic targets in dystonia based on recent preclinical animal studies and clinical trials investigating novel therapeutics.
Collapse
|
24
|
Gonzalez-Alegre P. Advances in molecular and cell biology of dystonia: Focus on torsinA. Neurobiol Dis 2019; 127:233-241. [DOI: 10.1016/j.nbd.2019.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/20/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
|
25
|
Pirio Richardson S, Jinnah HA. New approaches to discovering drugs that treat dystonia. Expert Opin Drug Discov 2019; 14:893-900. [PMID: 31159587 DOI: 10.1080/17460441.2019.1623785] [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] [Indexed: 02/08/2023]
Abstract
Introduction: Dystonia consists of involuntary movements, abnormal posturing, and pain. In adults, dystonia presents in a particular region of the body and causes significant disability due to pain as well as impairment in activities of daily living and employment. The current gold standard treatment, botulinum toxin (BoNT), has limitations - painful, frequent injections due to 'wearing off' of treatment effect; expense; and expected side effects like swallowing difficulty and weakness. There is a clear therapeutic gap in our current treatment options for dystonia and also a clear need for an effective novel treatment. Testing any novel treatment is complicated because most adults with focal dystonia are treated with BoNT. Areas covered: This review focuses on establishing the need for novel therapeutics. It also suggests potential leads from preclinical studies; and, discusses the issue of clinical trial readiness in the dystonia field. Expert opinion: Identifying a novel therapeutic intervention for dystonia patients faces two major challenges. The first is acknowledging the therapeutic gap that currently exists. Second, shifting some of our research aims in dystonia to clinical trial readiness is imperative if we are to be ready to test novel therapeutic agents.
Collapse
Affiliation(s)
- Sarah Pirio Richardson
- a Department of Neurology, University of New Mexico Health Sciences Center , Albuquerque , NM , USA.,b Neurology Service, New Mexico Veterans Affairs Health Care System , Albuquerque , NM , USA
| | - H A Jinnah
- c Departments of Neurology, Human Genetics & Pediatrics, Emory University School of Medicine , Atlanta , Georgia
| |
Collapse
|
26
|
Xu Y, Li Z. Imaging metabotropic glutamate receptor system: Application of positron emission tomography technology in drug development. Med Res Rev 2019; 39:1892-1922. [DOI: 10.1002/med.21566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Youwen Xu
- Independent Consultant and Contractor, Radiopharmaceutical Development, Validation and Bio-Application; Philadelphia Pennsylvania
| | - Zizhong Li
- Pharmaceutical Research and Development, SOFIE Biosciences; Somerset New Jersey
| |
Collapse
|
27
|
Dystonia: Are animal models relevant in therapeutics? Rev Neurol (Paris) 2018; 174:608-614. [DOI: 10.1016/j.neurol.2018.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023]
|
28
|
Beauvais G, Rodriguez-Losada N, Ying L, Zakirova Z, Watson JL, Readhead B, Gadue P, French DL, Ehrlich ME, Gonzalez-Alegre P. Exploring the Interaction Between eIF2α Dysregulation, Acute Endoplasmic Reticulum Stress and DYT1 Dystonia in the Mammalian Brain. Neuroscience 2018; 371:455-468. [PMID: 29289717 DOI: 10.1016/j.neuroscience.2017.12.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022]
|
29
|
Sun HJ, Chen D, Han Y, Zhou YB, Wang JJ, Chen Q, Li YH, Gao XY, Kang YM, Zhu GQ. Relaxin in paraventricular nucleus contributes to sympathetic overdrive and hypertension via PI3K-Akt pathway. Neuropharmacology 2016; 103:247-56. [DOI: 10.1016/j.neuropharm.2015.12.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/29/2015] [Accepted: 12/28/2015] [Indexed: 12/01/2022]
|
30
|
Metabotropic glutamate receptor 5 – a promising target in drug development and neuroimaging. Eur J Nucl Med Mol Imaging 2016; 43:1151-70. [DOI: 10.1007/s00259-015-3301-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
|
31
|
Dopamine and Its Actions in the Basal Ganglia System. INNOVATIONS IN COGNITIVE NEUROSCIENCE 2016. [DOI: 10.1007/978-3-319-42743-0_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
32
|
Rhes regulates dopamine D2 receptor transmission in striatal cholinergic interneurons. Neurobiol Dis 2015; 78:146-61. [PMID: 25818655 DOI: 10.1016/j.nbd.2015.03.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/12/2015] [Accepted: 03/17/2015] [Indexed: 11/22/2022] Open
Abstract
Ras homolog enriched in striatum (Rhes) is highly expressed in striatal medium spiny neurons (MSNs) of rodents. In the present study, we characterized the expression of Rhes mRNA across species, as well as its functional role in other striatal neuron subtypes. Double in situ hybridization analysis showed that Rhes transcript is selectively localized in striatal cholinergic interneurons (ChIs), but not in GABAergic parvalbumin- or in neuropeptide Y-positive cell populations. Rhes is closely linked to dopamine-dependent signaling. Therefore, we recorded ChIs activity in basal condition and following dopamine receptor activation. Surprisingly, instead of an expected dopamine D2 receptor (D2R)-mediated inhibition, we observed an aberrant excitatory response in ChIs from Rhes knockout mice. Conversely, the effect of D1R agonist on ChIs was less robust in Rhes mutants than in controls. Although Rhes deletion in mutants occurs throughout the striatum, we demonstrate that the D2R response is altered specifically in ChIs, since it was recorded in pharmacological isolation, and prevented either by intrapipette BAPTA or by GDP-β-S. Moreover, we show that blockade of Cav2.2 calcium channels prevented the abnormal D2R response. Finally, we found that the abnormal D2R activation in ChIs was rescued by selective PI3K inhibition thus suggesting that Rhes functionally modulates PI3K/Akt signaling pathway in these neurons. Our findings reveal that, besides its expression in MSNs, Rhes is localized also in striatal ChIs and, most importantly, lack of this G-protein, significantly alters D2R modulation of striatal cholinergic excitability.
Collapse
|