1
|
Rodriguez-Contreras D, García-Nafría J, Chan AE, Shinde U, Neve KA. Comparison of the function of two novel human dopamine D2 receptor variants identifies a likely mechanism for their pathogenicity. Biochem Pharmacol 2024; 228:116228. [PMID: 38643909 PMCID: PMC11410538 DOI: 10.1016/j.bcp.2024.116228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/29/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Two recently discovered DRD2 mutations, c.634A > T, p.Ile212Phe and c.1121T > G, p.Met374Arg, cause hyperkinetic movement disorders that have overlapping features but apparently differ in severity. The two known carriers of the Met374Arg variant had early childhood disease onset and more severe motor, cognitive, and neuropsychiatric deficits than any known carriers of the Ile212Phe variant, whose symptoms were first apparent in adolescence. Here, we evaluated if differences in the function of the two variants in cultured cells could explain differing pathogenicity. Both variants were expressed less abundantly than the wild type receptor and exhibited loss of agonist-induced arrestin binding, but differences in expression and arrestin binding between the variants were minor. Basal and agonist-induced activation of heterotrimeric Gi/o/z proteins, however, showed clear differences; agonists were generally more potent at Met374Arg than at the Ile212Phe or wild type variants. Furthermore, all Gα subtypes tested were constitutively activated more by Met374Arg than by Ile212Phe. Met374Arg produced greater constitutive inhibition of cyclic AMP accumulation than Ile212Phe or the wild type D2 receptor. Met374Arg and Ile212Phe were more sensitive to thermal inactivation than the wild type D2 receptor, as reported for other constitutively active receptors, but Ile212Phe was affected more than Met374Arg. Additional pharmacological characterization suggested that the mutations differentially affect the shape of the agonist binding pocket and the potency of dopamine, norepinephrine, and tyramine. Molecular dynamics simulations provided a structural rationale for enhanced constitutive activation and agonist potency. Enhanced constitutive and agonist-induced G protein-mediated signaling likely contributes to the pathogenicity of these novel variants.
Collapse
Affiliation(s)
- Dayana Rodriguez-Contreras
- Research Service, Veterans Affairs Portland Health Care System, and Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Javier García-Nafría
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Laboratory of Advanced Microscopy (LMA), University of Zaragoza, 50018, Zaragoza, Spain
| | - Amy E Chan
- Research Service, Veterans Affairs Portland Health Care System, and Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ujwal Shinde
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kim A Neve
- Research Service, Veterans Affairs Portland Health Care System, and Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
2
|
Yen YC, Li Y, Chen CL, Klose T, Watts VJ, Dessauer CW, Tesmer JJG. Structure of adenylyl cyclase 5 in complex with Gβγ offers insights into ADCY5-related dyskinesia. Nat Struct Mol Biol 2024; 31:1189-1197. [PMID: 38589608 PMCID: PMC11329361 DOI: 10.1038/s41594-024-01263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 03/04/2024] [Indexed: 04/10/2024]
Abstract
The nine different membrane-anchored adenylyl cyclase isoforms (AC1-9) in mammals are stimulated by the heterotrimeric G protein, Gαs, but their response to Gβγ regulation is isoform specific. In the present study, we report cryo-electron microscope structures of ligand-free AC5 in complex with Gβγ and a dimeric form of AC5 that could be involved in its regulation. Gβγ binds to a coiled-coil domain that links the AC transmembrane region to its catalytic core as well as to a region (C1b) that is known to be a hub for isoform-specific regulation. We confirmed the Gβγ interaction with both purified proteins and cell-based assays. Gain-of-function mutations in AC5 associated with human familial dyskinesia are located at the interface of AC5 with Gβγ and show reduced conditional activation by Gβγ, emphasizing the importance of the observed interaction for motor function in humans. We propose a molecular mechanism wherein Gβγ either prevents dimerization of AC5 or allosterically modulates the coiled-coil domain, and hence the catalytic core. As our mechanistic understanding of how individual AC isoforms are uniquely regulated is limited, studies such as this may provide new avenues for isoform-specific drug development.
Collapse
Affiliation(s)
- Yu-Chen Yen
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Yong Li
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chun-Liang Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Thomas Klose
- Purdue Cryo-EM Facility, Hockmeyer Hall for Structural Biology, Purdue University, West Lafayette, IN, USA
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - John J G Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
3
|
Spoto G, Ceraolo G, Butera A, Di Rosa G, Nicotera AG. Exploring the Genetic Landscape of Chorea in Infancy and Early Childhood: Implications for Diagnosis and Treatment. Curr Issues Mol Biol 2024; 46:5632-5654. [PMID: 38921008 PMCID: PMC11202702 DOI: 10.3390/cimb46060337] [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: 04/21/2024] [Revised: 06/01/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
Chorea is a hyperkinetic movement disorder frequently observed in the pediatric population, and, due to advancements in genetic techniques, an increasing number of genes have been associated with this disorder. In genetic conditions, chorea may be the primary feature of the disorder, or be part of a more complex phenotype characterized by epileptic encephalopathy or a multisystemic syndrome. Moreover, it can appear as a persistent disorder (chronic chorea) or have an episodic course (paroxysmal chorea). Managing chorea in childhood presents challenges due to its varied clinical presentation, often involving a spectrum of hyperkinetic movement disorders alongside neuropsychiatric and multisystemic manifestations. Furthermore, during infancy and early childhood, transient motor phenomena resembling chorea occurring due to the rapid nervous system development during this period can complicate the diagnosis. This review aims to provide an overview of the main genetic causes of pediatric chorea that may manifest during infancy and early childhood, focusing on peculiarities that can aid in differential diagnosis among different phenotypes and discussing possible treatment options.
Collapse
Affiliation(s)
- Giulia Spoto
- Unit of Child Neurology and Psychiatry, Department of Biomedical Sciences, Dental Sciences & Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Graziana Ceraolo
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Ambra Butera
- Unit of Child Neurology and Psychiatry, Department of Chemical, Biological, Farmaceutical & Environmental Science, University of Messina, 98125 Messina, Italy;
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Biomedical Sciences, Dental Sciences & Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Antonio Gennaro Nicotera
- Unit of Child Neurology and Psychiatry, Maternal-Infantile Department, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
4
|
Kanai Y. Molecular pathological approach to cancer epigenomics and its clinical application. Pathol Int 2024; 74:167-186. [PMID: 38482965 PMCID: PMC11551818 DOI: 10.1111/pin.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/15/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Careful microscopic observation of histopathological specimens, accumulation of large numbers of high-quality tissue specimens, and analysis of molecular pathology in relation to morphological features are considered to yield realistic data on the nature of multistage carcinogenesis. Since the morphological hallmark of cancer is disruption of the normal histological structure maintained through cell-cell adhesiveness and cellular polarity, attempts have been made to investigate abnormalities of the cadherin-catenin cell adhesion system in human cancer cells. It has been shown that the CDH1 tumor suppressor gene encoding E-cadherin is silenced by DNA methylation, suggesting that a "double hit" involving DNA methylation and loss of heterozygosity leads to carcinogenesis. Therefore, in the 1990s, we focused on epigenomic mechanisms, which until then had not received much attention. In chronic hepatitis and liver cirrhosis associated with hepatitis virus infection, DNA methylation abnormalities were found to occur frequently, being one of the earliest indications that such abnormalities are present even in precancerous tissue. Aberrant expression and splicing of DNA methyltransferases, such as DNMT1 and DNMT3B, was found to underlie the mechanism of DNA methylation alterations in various organs. The CpG island methylator phenotype in renal cell carcinoma was identified for the first time, and its therapeutic targets were identified by multilayer omics analysis. Furthermore, the DNA methylation profile of nonalcoholic steatohepatitis (NASH)-related hepatocellular carcinoma was clarified in groundbreaking studies. Since then, we have developed diagnostic markers for carcinogenesis risk in NASH patients and noninvasive diagnostic markers for upper urinary tract cancer, as well as developing a new high-performance liquid chromatography-based diagnostic system for DNA methylation diagnosis. Research on the cancer epigenome has revealed that DNA methylation alterations occur from the precancerous stage as a result of exposure to carcinogenic factors such as inflammation, smoking, and viral infections, and continuously contribute to multistage carcinogenesis through aberrant expression of cancer-related genes and genomic instability. DNA methylation alterations at the precancerous stages are inherited by or strengthened in cancers themselves and determine the clinicopathological aggressiveness of cancers as well as patient outcome. DNA methylation alterations have applications as biomarkers, and are expected to contribute to diagnosis, as well as preventive and preemptive medicine.
Collapse
Affiliation(s)
- Yae Kanai
- Department of PathologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
5
|
Makiuchi S, Tian Y, Fujimoto M, Kuramoto J, Tsuda N, Ojima H, Gotoh M, Hiraoka N, Yoshida T, Kanai Y, Arai E. DNA methylation alterations of ADCY5, MICAL2, and PLEKHG2 during the developmental stage of cryptogenic hepatocellular carcinoma. Hepatol Res 2024; 54:284-299. [PMID: 37906571 DOI: 10.1111/hepr.13984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
AIM The aim of this study was to clarify the significance of DNA methylation alterations of cryptogenic hepatocellular carcinomas (HCCs). METHODS Using the Infinium assay, we performed genome-wide DNA methylation analysis of 250 liver tissue samples, including noncancerous liver tissue (U-N) and corresponding cancerous tissue (U-T) from patients with cryptogenic HCC without a history of excessive alcohol use and hepatitis virus infection, and whose U-N samples showed no remarkable histological features (no microscopic evidence of simple steatosis, any form of hepatitis including nonalcoholic steatohepatitis, or liver cirrhosis). RESULTS We identified 3272 probes that showed significant differences of DNA methylation levels between U-N and normal liver tissue samples from patients without HCC, indicating that a distinct DNA methylation profile had already been established at the precancerous U-N stage. U-Ns have a DNA methylation profile differing from that of noncancerous liver tissue of patients with nonalcoholic steatohepatitis-related, viral hepatitis-related, and alcoholic liver disease-related HCCs. Such DNA methylation alterations in U-Ns were inherited by U-Ts. The U-Ns showed DNA methylation alteration of ADCY5, resulting in alteration of its mRNA expression, whereas noncancerous liver tissue of patients with nonalcoholic steatohepatitis-, viral hepatitis-, or alcoholic liver disease-related HCCs did not. DNA methylation levels of MICAL2 and PLEKHG2 in U-Ts were correlated with larger tumor diameter and portal vein involvement, respectively. CONCLUSIONS U-N-specific DNA hypermethylation of ADCY5 may have significance, even from the precancerous stage in liver showing no remarkable histological features. DNA hypomethylation of MICAL2 and PLEKHG2 may determine the clinicopathological features of cryptogenic HCC.
Collapse
Affiliation(s)
- Satomi Makiuchi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Ying Tian
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mao Fujimoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Junko Kuramoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Noboru Tsuda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Gotoh
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Teruhiko Yoshida
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Thomsen M, Lange LM, Zech M, Lohmann K. Genetics and Pathogenesis of Dystonia. ANNUAL REVIEW OF PATHOLOGY 2024; 19:99-131. [PMID: 37738511 DOI: 10.1146/annurev-pathmechdis-051122-110756] [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: 09/24/2023]
Abstract
Dystonia is a clinically and genetically highly heterogeneous neurological disorder characterized by abnormal movements and postures caused by involuntary sustained or intermittent muscle contractions. A number of groundbreaking genetic and molecular insights have recently been gained. While they enable genetic testing and counseling, their translation into new therapies is still limited. However, we are beginning to understand shared pathophysiological pathways and molecular mechanisms. It has become clear that dystonia results from a dysfunctional network involving the basal ganglia, cerebellum, thalamus, and cortex. On the molecular level, more than a handful of, often intertwined, pathways have been linked to pathogenic variants in dystonia genes, including gene transcription during neurodevelopment (e.g., KMT2B, THAP1), calcium homeostasis (e.g., ANO3, HPCA), striatal dopamine signaling (e.g., GNAL), endoplasmic reticulum stress response (e.g., EIF2AK2, PRKRA, TOR1A), autophagy (e.g., VPS16), and others. Thus, different forms of dystonia can be molecularly grouped, which may facilitate treatment development in the future.
Collapse
Affiliation(s)
- Mirja Thomsen
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| |
Collapse
|
7
|
Pontrucher A, Barth M, Ziegler A, Chao de la Barca JM, Mirebeau-Prunier D, Reynier P, Homedan C. Case report: Diagnosis of ADCY5-related dyskinesia explaining the entire phenotype in a patient with atypical citrullinemia type I. Front Neurol 2023; 14:1266686. [PMID: 38020658 PMCID: PMC10665474 DOI: 10.3389/fneur.2023.1266686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
In this case study, we report the case of a 13-year-old girl with citrullinemia type 1 (MIM #215700), an autosomal recessive inherited disorder of the urea cycle, which was confirmed by the identification of a homozygous pathogenic variant in the argininosuccinate synthetase 1 (ASS1) gene. However, the patient presented abnormal hyperkinetic movements with global developmental delay and clinical signs that were not fully consistent with those of citrullinemia type 1 or with those of her siblings with isolated citrullinemia type 1. Exome sequencing showed the presence of a de novo heterozygous pathogenic variant in the adenylate cyclase type 5 (ADCY5) gene. The variant confirmed the overlap with the so-called ADCY5-related dyskinesia with orofacial involvement, which is autosomal dominant (MIM #606703), a disorder disrupting the enzymatic conversion of adenosine triphosphate (ATP) to cyclic adenosine monophosphate (cAMP). In addition to the citrullinemia-related low-protein diet and arginine supplementation, the identification of this second disease led to the introduction of a treatment with caffeine, which considerably improved the dyskinesia neurological picture. In conclusion, this case highlights the importance of clinical-biological confrontation for the interpretation of genetic variants, as one hereditary metabolic disease may hide another with therapeutic consequences. Summary This article reports the misleading superposition of two inherited metabolic diseases, showing the importance of clinical-biological confrontation in the interpretation of genetic variants.
Collapse
Affiliation(s)
- Audrey Pontrucher
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire, Angers, France
| | - Magalie Barth
- Service de Génétique, Centre Hospitalier Universitaire, Angers, France
| | - Alban Ziegler
- Service de Génétique, Centre Hospitalier Universitaire, Angers, France
- Service de Génétique, CRMR AnDDI-Rares, CHU Reims, Reims, France
| | | | | | - Pascal Reynier
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire, Angers, France
| | - Chadi Homedan
- Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire, Angers, France
| |
Collapse
|
8
|
Menon PJ, Nilles C, Silveira‐Moriyama L, Yuan R, de Gusmao CM, Münchau A, Carecchio M, Grossman S, Grossman G, Méneret A, Roze E, Pringsheim T. Scoping Review on ADCY5-Related Movement Disorders. Mov Disord Clin Pract 2023; 10:1048-1059. [PMID: 37476318 PMCID: PMC10354615 DOI: 10.1002/mdc3.13796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/29/2023] [Accepted: 05/04/2023] [Indexed: 07/22/2023] Open
Abstract
Background Adenylyl cyclase 5 (ADCY5)-related movement disorder (ADCY5-RMD) is a rare, childhood-onset disease resulting from pathogenic variants in the ADCY5 gene. The clinical features, diagnostic options, natural history, and treatments for this disease are poorly characterized and have never been established through a structured approach. Objective This scoping review attempts to summarize all available clinical literature on ADCY5-RMD. Methods Eighty-seven articles were selected for inclusion in this scoping review. The majority of articles identified were case reports or case series. Results These articles demonstrate that patients with ADCY5-RMD suffer from permanent and/ or paroxysmal hyperkinetic movements. The paroxysmal episodes can be worsened by environmental triggers, in particular the sleep-wake transition phase in the early morning. Occurrence of nocturnal paroxysmal dyskinesias and perioral twitches are highly suggestive of the diagnosis when present. In the majority of patients intellectual capacity is preserved. ADCY5-RMD is considered a non-progressive disorder, with inter-individual variations in evolution with aging. Somatic mosaicism, mode of inheritance and the location of the mutation within the protein can influence phenotype. Conclusions The current evidence for therapeutic options for ADCY5-RMD is limited: caffeine, benzodiazepines and deep brain stimulation have been consistently reported to be useful in case reports and case series.
Collapse
Affiliation(s)
- Poornima Jayadev Menon
- Sorbonne University, APHP—Salpêtrière Hospital, CNRS, INSERM, Paris Brain InstituteParisFrance
- School of Postgraduate StudiesRoyal College of Surgeons in IrelandDublinIreland
| | - Christelle Nilles
- Department of Clinical NeurosciencesUniversity of CalgaryCalgaryABCanada
| | | | - Ruiyi Yuan
- Sorbonne University, APHP—Salpêtrière Hospital, CNRS, INSERM, Paris Brain InstituteParisFrance
| | - Claudio M. de Gusmao
- Department of NeurologyUniversity of Campinas (UNICAMP)CampinasBrazil
- Boston Children's HospitalBostonMAUSA
| | | | - Miryam Carecchio
- Center for the Study of Neurodegeneration (CESNE) and Department of NeuroscienceUniversity of PaduaPaduaItaly
| | | | | | - Aurélie Méneret
- Sorbonne University, APHP—Salpêtrière Hospital, CNRS, INSERM, Paris Brain InstituteParisFrance
| | - Emmanuel Roze
- Sorbonne University, APHP—Salpêtrière Hospital, CNRS, INSERM, Paris Brain InstituteParisFrance
| | - Tamara Pringsheim
- Department of Clinical NeurosciencesUniversity of CalgaryCalgaryABCanada
| |
Collapse
|
9
|
Yen YC, Li Y, Chen CL, Klose T, Watts VJ, Dessauer CW, Tesmer JJG. Isoform Specific Regulation of Adenylyl Cyclase 5 by Gβγ. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539090. [PMID: 37205557 PMCID: PMC10187219 DOI: 10.1101/2023.05.02.539090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The nine different membrane-anchored adenylyl cyclase isoforms (AC1-9) in mammals are stimulated by the heterotrimeric G protein Gαs, but their response to Gβγ regulation is isoform-specific. For example, AC5 is conditionally activated by Gβγ. Here, we report cryo-EM structures of ligand-free AC5 in complex with Gβγ and of a dimeric form of AC5 that could be involved in its regulation. Gβγ binds to a coiled-coil domain that links the AC transmembrane region to its catalytic core as well as to a region (C1b) that is known to be a hub for isoform-specific regulation. We confirmed the Gβγ interaction with both purified proteins and cell-based assays. The interface with Gβγ involves AC5 residues that are subject to gain-of-function mutations in humans with familial dyskinesia, indicating that the observed interaction is important for motor function. A molecular mechanism wherein Gβγ either prevents dimerization of AC5 or allosterically modulates the coiled-coil domain, and hence the catalytic core, is proposed. Because our mechanistic understanding of how individual AC isoforms are uniquely regulated is limited, studies such as this may provide new avenues for isoform-specific drug development.
Collapse
Affiliation(s)
- Yu-Chen Yen
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Yong Li
- Department Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chun-Liang Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Thomas Klose
- Purdue CryoEM Facility, Suite 171, Hockmeyer Hall for Structural Biology, Purdue University, West Lafayette, IN, USA
| | - Val J Watts
- Department of Molecular Pharmacology and Medicinal Chemistry, Purdue University, West Lafayette, IN, USA
| | - Carmen W Dessauer
- Department Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - John J. G. Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Department of Molecular Pharmacology and Medicinal Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
10
|
Chen Z, Antoni FA. Human adenylyl cyclase 9 is auto-stimulated by its isoform-specific C-terminal domain. Life Sci Alliance 2023; 6:e202201791. [PMID: 36657828 PMCID: PMC9873982 DOI: 10.26508/lsa.202201791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Human transmembrane adenylyl cyclase 9 (AC9) is not regulated by heterotrimeric G proteins. Key to the resistance to stimulation by Gs-coupled receptors (GsRs) is auto-inhibition by the COOH-terminal domain (C2b). The present study investigated the role of the C2b domain in the regulation of cyclic AMP production by AC9 in HEK293FT cells expressing the GloSensor22F cyclic AMP-reporter protein. Surprisingly, we found C2b to be essential for sustaining the basal output of cyclic AMP by AC9. A human mutation (E326D) in the parallel coiled-coil formed by the signalling helices of AC9 dramatically increased basal activity, which was also dependent on the C2b domain. Intriguingly, the same mutation enabled stimulation of AC9 by GsRs. In summary, auto-regulation by the C2b domain of AC9 sustains its basal activity and quenches activation by GsR. Thus, AC9 appears to be tailored to support constitutive activation of cyclic AMP effector systems. A switch from this paradigm to stimulation by GsRs may be occasioned by conformational changes at the coiled-coil or removal of the C2b domain.
Collapse
Affiliation(s)
- Zhihao Chen
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Ferenc A Antoni
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Rodriguez-Contreras D, Gong S, Lebowitz JJ, Fedorov LM, Asad N, Dore TM, Phillips TJ, Ford CP, Williams JT, Neve KA. Gait Abnormalities and Aberrant D2 Receptor Expression and Signaling in Mice Carrying the Human Pathogenic Mutation DRD2I212F. Mol Pharmacol 2023; 103:188-198. [PMID: 36456191 PMCID: PMC11033946 DOI: 10.1124/molpharm.122.000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/21/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
A dopamine D2 receptor mutation was recently identified in a family with a novel hyperkinetic movement disorder. That allelic variant D2-I212F is a constitutively active and G protein-biased receptor. We now describe mice engineered using CRISPR-Cas9-mediated gene editing technology to carry the D2-I212F variant. Drd2I212F mice exhibited gait abnormalities resembling those in other mouse models of chorea and/or dystonia and had striatal D2 receptor expression that was decreased approximately 30% per Drd2I212F allele. Electrically evoked inhibitory postsynaptic conductances in midbrain dopamine neurons and striatum from Drd2I212F mice, caused by G protein activation of potassium channels, exhibited slow kinetics (e.g., approximately four- to sixfold slower decay) compared with Drd2 +/+ mice. Current decay initiated by photolytic release of the D2 antagonist sulpiride from CyHQ-sulpiride was also ∼fourfold slower in midbrain slices from Drd2I212F mice than Drd2 +/+ mice. Furthermore, in contrast to Drd2 +/+ mice, in which dopamine is several-fold more potent at neurons in the nucleus accumbens than in the dorsal striatum, reflecting activation of Gα o versus Gα i, dopamine had similar potencies in those two brain regions of Drd2I212F mice. Repeated cocaine treatment, which decreases dopamine potency in the nucleus accumbens of Drd2 +/+ mice, had no effect on dopamine potency in Drd2 I212F mice. The results demonstrate the pathogenicity of the D2-I212F mutation and the utility of this mouse model for investigating the role of pathogenic DRD2 variants in early-onset hyperkinetic movement disorders. SIGNIFICANCE STATEMENT: The first dopamine receptor mutation to cause a movement disorder, D2-I212F, was recently identified. The mutation makes receptor activation of G protein-mediated signaling more efficient. To confirm the pathogenesis of D2-I212F, this study reports that mice carrying this mutation have gait abnormalities consistent with the clinical phenotype. The mutation also profoundly alters D2 receptor expression and function in vivo. This mouse model will be useful for further characterization of the mutant receptor and for evaluation of potential therapeutic drugs.
Collapse
Affiliation(s)
- Dayana Rodriguez-Contreras
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Sheng Gong
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Joseph J Lebowitz
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Lev M Fedorov
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Naeem Asad
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Timothy M Dore
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Tamara J Phillips
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Christopher P Ford
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - John T Williams
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Kim A Neve
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| |
Collapse
|
12
|
Tänzler D, Kipping M, Lederer M, Günther WF, Arlt C, Hüttelmaier S, Merkenschlager A, Sinz A. Effects of theophylline on ADCY5 activation-From cellular studies to improved therapeutic options for ADCY5-related dyskinesia patients. PLoS One 2023; 18:e0282593. [PMID: 36867608 PMCID: PMC9983822 DOI: 10.1371/journal.pone.0282593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
We show the effects of the three purine derivatives, caffeine, theophylline, and istradefylline, on cAMP production by adenylyl cyclase 5 (ADCY5)-overexpressing cell lines. A comparison of cAMP levels was performed for ADCY5 wild-type and R418W mutant cells. ADCY5-catalyzed cAMP production was reduced with all three purine derivatives, while the most pronounced effects on cAMP reduction were observed for ADCY5 R418W mutant cells. The gain-of-function ADCY5 R418W mutant is characterized by an increased catalytic activity resulting in elevated cAMP levels that cause kinetic disorders or dyskinesia in patients. Based on our findings in ADCY5 cells, a slow-release formulation of theophylline was administered to a preschool-aged patient with ADCY5-related dyskinesia. A striking improvement of symptoms was observed, outperforming the effects of caffeine that had previously been administered to the same patient. We suggest considering theophylline as an alternative therapeutic option to treat ADCY5-related dyskinesia in patients.
Collapse
Affiliation(s)
- Dirk Tänzler
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
| | - Marc Kipping
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
| | - Marcell Lederer
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Saxony-Anhalt, Germany
| | - Wiebke F. Günther
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
| | - Christian Arlt
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Saxony-Anhalt, Germany
| | - Andreas Merkenschlager
- Department of Neuropediatrics, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Saxony, Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- Center for Structural Mass Spectrometry, Martin Luther University Halle-Wittenberg, Halle (Saale), Saxony-Anhalt, Germany
- * E-mail:
| |
Collapse
|
13
|
ADCY5 gene mutation: a case report. Neurol Sci 2022; 43:6947-6950. [DOI: 10.1007/s10072-022-06394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
|
14
|
Méneret A, Mohammad SS, Cif L, Doummar D, DeGusmao C, Anheim M, Barth M, Damier P, Demonceau N, Friedman J, Gallea C, Gras D, Gurgel-Giannetti J, Innes EA, Necpál J, Riant F, Sagnes S, Sarret C, Seliverstov Y, Paramanandam V, Shetty K, Tranchant C, Doulazmi M, Vidailhet M, Pringsheim T, Roze E. Efficacy of Caffeine in ADCY5-Related Dyskinesia: A Retrospective Study. Mov Disord 2022; 37:1294-1298. [PMID: 35384065 DOI: 10.1002/mds.29006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/08/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND ADCY5-related dyskinesia is characterized by early-onset movement disorders. There is currently no validated treatment, but anecdotal clinical reports and biological hypotheses suggest efficacy of caffeine. OBJECTIVE The aim is to obtain further insight into the efficacy and safety of caffeine in patients with ADCY5-related dyskinesia. METHODS A retrospective study was conducted worldwide in 30 patients with a proven ADCY5 mutation who had tried or were taking caffeine for dyskinesia. Disease characteristics and treatment responses were assessed through a questionnaire. RESULTS Caffeine was overall well tolerated, even in children, and 87% of patients reported a clear improvement. Caffeine reduced the frequency and duration of paroxysmal movement disorders but also improved baseline movement disorders and some other motor and nonmotor features, with consistent quality-of-life improvement. Three patients reported worsening. CONCLUSION Our findings suggest that caffeine should be considered as a first-line therapeutic option in ADCY5-related dyskinesia. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Aurélie Méneret
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| | - Shekeeb S Mohammad
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Laura Cif
- Département de Neurochirurgie, Hôpital Gui de Chauliac, Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Diane Doummar
- Service de Neuropédiatrie-Pathologie du développement, centre de référence mouvements anormaux enfant, Hôpital Trousseau AP-HP.SU, FHU I2D2, Sorbonne Université, Paris, France
| | | | - Mathieu Anheim
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - Philippe Damier
- CHU de Nantes, INSERM, CIC 1314, Hôpital Laennec, Nantes, France
| | | | - Jennifer Friedman
- Departments of Neurosciences and Pediatrics, University of California San Diego, La Jolla, California, USA.,Division of Neurology, Rady Children's Hospital, San Diego, California, USA.,Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Cécile Gallea
- Sorbonne University, INSERM, CNRS, Paris Brain Institute, Paris, France
| | - Domitille Gras
- U1141 Neurodiderot, équipe 5 inDev, Inserm, CEA, UP, UNIACTNeurospin, Joliot, DRF, CEA, Saclay, France
| | | | - Emily A Innes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia.,University of Notre Dame Australia, School of Medicine, Sydney, NSW, Australia
| | - Ján Necpál
- Department of Neurology, Zvolen Hospital, Zvolen, Slovakia
| | - Florence Riant
- Service de Génétique Moléculaire, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sandrine Sagnes
- Délégation à la Recherche Clinique et à l'Innovation-DRCI (Clinical Research and Innovation Department) and URC (Clinical Research Unit) GH Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Catherine Sarret
- Service de pédiatrie, hôpital Estaing, Centre hospitalier universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Yury Seliverstov
- Research Center of Neurology, Moscow, Russia.,Kazaryan Clinic of Epileptology and Neurology, Moscow, Russia
| | | | - Kuldeep Shetty
- Department of Neurology, Mazumdar Shaw Medical Center, Bangalore, India
| | - Christine Tranchant
- Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM-U964/CNRS-UMR7104/Université de Strasbourg, Illkirch-Graffenstaden, France.,Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Mohamed Doulazmi
- Adaptation Biologique et Vieillissement, Institut de Biologie Paris Seine, Sorbonne University, CNRS, Paris, France
| | - Marie Vidailhet
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| | - Tamara Pringsheim
- Department of Clinical Neurosciences, Psychiatry, Pediatrics and Community Health Sciences, University of Calgary, Calgary, Canada
| | - Emmanuel Roze
- Inserm U1127, CNRS UMR7225, UM75, Paris Brain Institute, Assistance Publique-Hôpitaux de Paris, DMU Neurosciences, Sorbonne University, Paris, France
| |
Collapse
|
15
|
Ostrom KF, LaVigne JE, Brust TF, Seifert R, Dessauer CW, Watts VJ, Ostrom RS. Physiological roles of mammalian transmembrane adenylyl cyclase isoforms. Physiol Rev 2022; 102:815-857. [PMID: 34698552 PMCID: PMC8759965 DOI: 10.1152/physrev.00013.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Adenylyl cyclases (ACs) catalyze the conversion of ATP to the ubiquitous second messenger cAMP. Mammals possess nine isoforms of transmembrane ACs, dubbed AC1-9, that serve as major effector enzymes of G protein-coupled receptors (GPCRs). The transmembrane ACs display varying expression patterns across tissues, giving the potential for them to have a wide array of physiological roles. Cells express multiple AC isoforms, implying that ACs have redundant functions. Furthermore, all transmembrane ACs are activated by Gαs, so it was long assumed that all ACs are activated by Gαs-coupled GPCRs. AC isoforms partition to different microdomains of the plasma membrane and form prearranged signaling complexes with specific GPCRs that contribute to cAMP signaling compartments. This compartmentation allows for a diversity of cellular and physiological responses by enabling unique signaling events to be triggered by different pools of cAMP. Isoform-specific pharmacological activators or inhibitors are lacking for most ACs, making knockdown and overexpression the primary tools for examining the physiological roles of a given isoform. Much progress has been made in understanding the physiological effects mediated through individual transmembrane ACs. GPCR-AC-cAMP signaling pathways play significant roles in regulating functions of every cell and tissue, so understanding each AC isoform's role holds potential for uncovering new approaches for treating a vast array of pathophysiological conditions.
Collapse
Affiliation(s)
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Tarsis F Brust
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| |
Collapse
|
16
|
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
|
17
|
Erro R, Mencacci NE, Bhatia KP. The Emerging Role of Phosphodiesterases in Movement Disorders. Mov Disord 2021; 36:2225-2243. [PMID: 34155691 PMCID: PMC8596847 DOI: 10.1002/mds.28686] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclic nucleotide phosphodiesterase (PDE) enzymes catalyze the hydrolysis and inactivation of the cyclic nucleotides cyclic adenosine monophosphate and cyclic guanosine monophosphate, which act as intracellular second messengers for many signal transduction pathways in the central nervous system. Several classes of PDE enzymes with specific tissue distributions and cyclic nucleotide selectivity are highly expressed in brain regions involved in cognitive and motor functions, which are known to be implicated in neurodegenerative diseases, such as Parkinson's disease and Huntington's disease. The indication that PDEs are intimately involved in the pathophysiology of different movement disorders further stems from recent discoveries that mutations in genes encoding different PDEs, including PDE2A, PDE8B, and PDE10A, are responsible for rare forms of monogenic parkinsonism and chorea. We here aim to provide a translational overview of the preclinical and clinical data on PDEs, the role of which is emerging in the field of movement disorders, offering a novel venue for a better understanding of their pathophysiology. Modulating cyclic nucleotide signaling, by either acting on their synthesis or on their degradation, represents a promising area for development of novel therapeutic approaches. The study of PDE mutations linked to monogenic movement disorders offers the opportunity of better understanding the role of PDEs in disease pathogenesis, a necessary step to successfully benefit the treatment of both hyperkinetic and hypokinetic movement disorders. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Niccoló E Mencacci
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
18
|
Riluzole Administration to Rats with Levodopa-Induced Dyskinesia Leads to Loss of DNA Methylation in Neuronal Genes. Cells 2021; 10:cells10061442. [PMID: 34207710 PMCID: PMC8228416 DOI: 10.3390/cells10061442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Dyskinesias are characterized by abnormal repetitive involuntary movements due to dysfunctional neuronal activity. Although levodopa-induced dyskinesia, characterized by tic-like abnormal involuntary movements, has no clinical treatment for Parkinson’s disease patients, animal studies indicate that Riluzole, which interferes with glutamatergic neurotransmission, can improve the phenotype. The rat model of Levodopa-Induced Dyskinesia is a unilateral lesion with 6-hydroxydopamine in the medial forebrain bundle, followed by the repeated administration of levodopa. The molecular pathomechanism of Levodopa-Induced Dyskinesia is still not deciphered; however, the implication of epigenetic mechanisms was suggested. In this study, we investigated the striatum for DNA methylation alterations under chronic levodopa treatment with or without co-treatment with Riluzole. Our data show that the lesioned and contralateral striata have nearly identical DNA methylation profiles. Chronic levodopa and levodopa + Riluzole treatments led to DNA methylation loss, particularly outside of promoters, in gene bodies and CpG poor regions. We observed that several genes involved in the Levodopa-Induced Dyskinesia underwent methylation changes. Furthermore, the Riluzole co-treatment, which improved the phenotype, pinpointed specific methylation targets, with a more than 20% methylation difference relative to levodopa treatment alone. These findings indicate potential new druggable targets for Levodopa-Induced Dyskinesia.
Collapse
|
19
|
Nosadini M, D'Onofrio G, Pelizza MF, Luisi C, Padrin D, Baggio L, Zorzi GS, Toldo I, Sartori S. [Not Available]. Neuropediatrics 2021; 52:208-211. [PMID: 33374026 DOI: 10.1055/s-0040-1721685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Abstract
Background Mutations in the adenylate cyclase 5 (ADCY5) gene are associated with childhood-onset paroxysmal dyskinesia.
Methods We report a new video-documented case of pediatric ADCY5-related dyskinesia with de novo ADCY5 mutation.
Results A boy born to nonconsanguineous parents after an uneventful pregnancy had developmental delay and hypotonia. At the age of 7 months, he presented with paroxysmal jerky–choreic–dystonic involuntary movements in wakefulness involving limbs, trunk, and face, exacerbated by emotional stimuli. These episodes gradually worsened in duration and frequency: at the age of 2.5 years, they occurred up to six times per day, and appeared also during sleep in prolonged bouts; the boy also had basal choreoathetoid–dystonic movements, hyperactivity, paraparetic–ataxic gait, generalized hypotonia with brisk tendon reflexes, drooling, and language delay with intellectual disability. Brain magnetic resonance imaging, electroencephalogram, electromyogram, eye review, metabolic investigations, oligoclonal bands, and autoantibodies were normal. Extensive genetic testing had not let to a diagnosis, until a heterozygous de novo mutation c.1252C > T (p.Arg418Trp) was identified in the ADCY5 gene. Clonazepam had partial effectiveness. The boy walked at the age of 3.5 years. At the age of 5 years, the paroxysmal movement disorder has slightly improved.
Conclusion ADCY5 mutations should be considered among the differential diagnoses of early-onset paroxysmal choreic–athetosic–myoclonic–dystonic movement disorder involving limbs, trunk, and face, in patients with global neurological impairment with hypotonia and developmental delay. Facial dyskinesias and exacerbation by drowsiness/sleep and emotional stimuli are important clues that may allow a timely recognition of the disorder and avoidance of unnecessary diagnostic investigations.
Collapse
Affiliation(s)
- Margherita Nosadini
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Gianluca D'Onofrio
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Maria Federica Pelizza
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Concetta Luisi
- Department of Neurosciences, Neurological Section, University of Padova, Padova, Italy
| | - Davide Padrin
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Laura Baggio
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | | | - Irene Toldo
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| | - Stefano Sartori
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, Padua, Italy
| |
Collapse
|
20
|
Ferrini A, Steel D, Barwick K, Kurian MA. An Update on the Phenotype, Genotype and Neurobiology of ADCY5-Related Disease. Mov Disord 2021; 36:1104-1114. [PMID: 33934385 DOI: 10.1002/mds.28495] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/23/2020] [Accepted: 12/21/2020] [Indexed: 01/11/2023] Open
Abstract
Adenylyl cyclase 5 (ADCY5)-related phenotypes comprise an expanding disease continuum, but much remains to be understood about the underlying pathogenic mechanisms of the disease. ADCY5-related disease comprises a spectrum of hyperkinetic disorders involving chorea, myoclonus, and/or dystonia, often with paroxysmal exacerbations. Hypotonia, developmental delay, and intellectual disability may be present. The causative gene encodes adenylyl cyclase, the enzyme responsible for the conversion of adenosine triphosphate (ATP) to cyclic adenosine-3',5'-monophosphate (cAMP). cAMP is a second messenger that exerts a wide variety of effects via several intracellular signaling pathways. ADCY5 is the most commonly expressed isoform of adenylyl cyclase in medium spiny neurons (MSNs) of the striatum, and it integrates and controls dopaminergic signaling. Through cAMP pathway, ADCY5 is a key regulator of the cortical and thalamic signaling that control initiation of voluntary movements and prevention of involuntary movements. Gain-of-function mutations in ADCY5 have been recently linked to a rare genetic disorder called ADCY5-related dyskinesia, where dysregulation of the cAMP pathway leads to reduced inhibitory activity and involuntary hyperkinetic movements. Here, we present an update on the neurobiology of ADCY5, together with a detailed overview of the reported clinical phenotypes and genotypes. Although a range of therapeutic approaches has been trialed, there are currently no disease-modifying treatments. Improved in vitro and in vivo laboratory models will no doubt increase our understanding of the pathogenesis of this rare genetic movement disorder, which will improve diagnosis, and also facilitate the development of precision medicine approaches for this, and other forms of hyperkinesia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Arianna Ferrini
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London, United Kingdom
| | - Dora Steel
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Katy Barwick
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London, United Kingdom
| | - Manju A Kurian
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London, United Kingdom
| |
Collapse
|
21
|
Gonzalez-Latapi P, Marotta N, Mencacci NE. Emerging and converging molecular mechanisms in dystonia. J Neural Transm (Vienna) 2021; 128:483-498. [DOI: 10.1007/s00702-020-02290-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023]
|
22
|
Guo Q, Li B, Bao C, Li Y, Cao Y, Wang C, Wu W. Bletilla striata
Polysaccharides Improve Hemostatic, Antiinflammatory Efficacy, and Platelet Aggregation in Gingivitis Rat Model. STARCH-STARKE 2020. [DOI: 10.1002/star.202000185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Qing Guo
- Department of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Bailei Li
- Department of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Chunling Bao
- Shanghai Sixth People's Hospital East Campus Shanghai 201306 China
| | - Yixia Li
- Nanjing Baiyun Chemical Environmental Monitoring Co., Ltd. Jiangsu 210047 China
| | - Yuling Cao
- Department of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Chunxiao Wang
- Department of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Wenhui Wu
- Department of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| |
Collapse
|
23
|
Delorme C, Giron C, Bendetowicz D, Méneret A, Mariani LL, Roze E. Current challenges in the pathophysiology, diagnosis, and treatment of paroxysmal movement disorders. Expert Rev Neurother 2020; 21:81-97. [PMID: 33089715 DOI: 10.1080/14737175.2021.1840978] [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] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Paroxysmal movement disorders mostly comprise paroxysmal dyskinesia and episodic ataxia, and can be the consequence of a genetic disorder or symptomatic of an acquired disease. AREAS COVERED In this review, the authors focused on certain hot-topic issues in the field: the respective contribution of the cerebellum and striatum to the generation of paroxysmal dyskinesia, the importance of striatal cAMP turnover in the pathogenesis of paroxysmal dyskinesia, the treatable causes of paroxysmal movement disorders not to be missed, with a special emphasis on the treatment strategy to bypass the glucose transport defect in paroxysmal movement disorders due to GLUT1 deficiency, and functional paroxysmal movement disorders. EXPERT OPINION Treatment of genetic causes of paroxysmal movement disorders is evolving towards precision medicine with targeted gene-specific therapy. Alteration of the cerebellar output and modulation of the striatal cAMP turnover offer new perspectives for experimental therapeutics, at least for paroxysmal movement disorders due to selected causes. Further characterization of cell-specific molecular pathways or network dysfunctions that are critically involved in the pathogenesis of paroxysmal movement disorders will likely result in the identification of new biomarkers and testing of innovative-targeted therapeutics.
Collapse
Affiliation(s)
- Cécile Delorme
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France
| | - Camille Giron
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France
| | - David Bendetowicz
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France.,Inserm U 1127, CNRS UMR 7225- Institut du cerveau (ICM), Sorbonne Université , Paris, France
| | - Aurélie Méneret
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France.,Inserm U 1127, CNRS UMR 7225- Institut du cerveau (ICM), Sorbonne Université , Paris, France
| | - Louise-Laure Mariani
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France.,Inserm U 1127, CNRS UMR 7225- Institut du cerveau (ICM), Sorbonne Université , Paris, France
| | - Emmanuel Roze
- Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière , Paris, France.,Inserm U 1127, CNRS UMR 7225- Institut du cerveau (ICM), Sorbonne Université , Paris, France
| |
Collapse
|
24
|
Baizabal-Carvallo JF, Cardoso F. Chorea in children: etiology, diagnostic approach and management. J Neural Transm (Vienna) 2020; 127:1323-1342. [DOI: 10.1007/s00702-020-02238-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/01/2020] [Indexed: 01/07/2023]
|
25
|
Clinical and Genetic Overview of Paroxysmal Movement Disorders and Episodic Ataxias. Int J Mol Sci 2020; 21:ijms21103603. [PMID: 32443735 PMCID: PMC7279391 DOI: 10.3390/ijms21103603] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Paroxysmal movement disorders (PMDs) are rare neurological diseases typically manifesting with intermittent attacks of abnormal involuntary movements. Two main categories of PMDs are recognized based on the phenomenology: Paroxysmal dyskinesias (PxDs) are characterized by transient episodes hyperkinetic movement disorders, while attacks of cerebellar dysfunction are the hallmark of episodic ataxias (EAs). From an etiological point of view, both primary (genetic) and secondary (acquired) causes of PMDs are known. Recognition and diagnosis of PMDs is based on personal and familial medical history, physical examination, detailed reconstruction of ictal phenomenology, neuroimaging, and genetic analysis. Neurophysiological or laboratory tests are reserved for selected cases. Genetic knowledge of PMDs has been largely incremented by the advent of next generation sequencing (NGS) methodologies. The wide number of genes involved in the pathogenesis of PMDs reflects a high complexity of molecular bases of neurotransmission in cerebellar and basal ganglia circuits. In consideration of the broad genetic and phenotypic heterogeneity, a NGS approach by targeted panel for movement disorders, clinical or whole exome sequencing should be preferred, whenever possible, to a single gene approach, in order to increase diagnostic rate. This review is focused on clinical and genetic features of PMDs with the aim to (1) help clinicians to recognize, diagnose and treat patients with PMDs as well as to (2) provide an overview of genes and molecular mechanisms underlying these intriguing neurogenetic disorders.
Collapse
|
26
|
The chilling of adenylyl cyclase 9 and its translational potential. Cell Signal 2020; 70:109589. [PMID: 32105777 DOI: 10.1016/j.cellsig.2020.109589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/21/2020] [Accepted: 02/23/2020] [Indexed: 12/26/2022]
Abstract
A recent break-through paper has revealed for the first time the high-resolution, three-dimensional structure of a mammalian trans-membrane adenylyl cyclase (tmAC) obtained by cryo-electronmicroscopy (cryo-EM). Reporting the structure of adenylyl cyclase 9 (AC9) in complex with activated Gsα, the cryo-EM study revealed that AC9 has three functionally interlinked, yet structurally distinct domains. The array of the twelve transmembrane helices is connected to the cytosolic catalytic core by two helical segments that are stabilized through the formation of a parallel coiled-coil. Surprisingly, in the presence of Gsα, the isoform-specific carboxyl-terminal tail of AC9 occludes the forskolin- as well as the active substrate-sites, resulting in marked autoinhibition of the enzyme. As AC9 has the lowest primary sequence homology with the eight further mammalian tmAC paralogues, it appears to be the best candidate for selective pharmacologic targeting. This is now closer to reality as the structural insight provided by the cryo-EM study indicates that all of the three structural domains are potential targets for bioactive agents. The present paper summarizes for molecular physiologists and pharmacologists what is known about the biological role of AC9, considers the potential modes of physiologic regulation, as well as pharmacologic targeting on the basis of the high-resolution cryo-EM structure. The translational potential of AC9 is considered upon highlighting the current state of genome-wide association screens, and the corresponding experimental evidence. Overall, whilst the high- resolution structure presents unique opportunities for the full understanding of the control of AC9, the data on the biological role of the enzyme and its translational potential are far from complete, and require extensive further study.
Collapse
|
27
|
Chen D, Latimer CS, Spencer M, Karna P, Gonzalez‐Cuyar LF, Davis MY, Keene CD, Bird TD, Raskind WH. Hyperphosphorylated Tau, Increased Adenylate Cyclase 5 (ADCY5) Immunoreactivity, but No Neuronal Loss in ADCY5-Dyskinesia. Mov Disord Clin Pract 2020; 7:70-77. [PMID: 31970214 PMCID: PMC6962666 DOI: 10.1002/mdc3.12873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adenylate cyclase 5 (ADCY5)-related dyskinesia is a childhood-onset movement disorder. Manifestations vary in frequency and severity and may include chorea, tremor, dystonia, facial twitches, myoclonus, axial hypotonia, and limb hypertonia. Psychosis is likely part of the broader spectrum. ADCY5 is widely expressed in the brain, especially in the striatum. Previous reports of brain autopsies of 2 subjects with likely ADCY5-dyskinesia were limited by the absence of a molecular diagnosis. In 1 case, normal gross pathology was reported. In the other case, ADCY5 expression was not examined and neuropathological findings were confounded by age and comorbidities. OBJECTIVES To examine ADCY5 expression and neuropathological changes in ADCY5-dyskinesia. METHODS An extensive brain autopsy, including immunohistochemical analyses with antibodies to paired helical filament tau, α-synuclein, amyloid-β, microtubule-associated protein 2, and ADCY5, was performed. RESULTS The patient, with a p.M1029K ADCY5 variant, had severe dyskinesias from early childhood, later recurrent episodes of psychosis, and died at age 46. Gross pathology was unremarkable, but we detected increased immunoreactivity for ADCY5 in neurons in multiple brain regions. Despite no history of brain trauma to suggest chronic traumatic encephalopathy, we found tau deposits in the deep cortical sulci, midbrain, and hippocampus with minimal amyloid pathology and no Lewy bodies. CONCLUSIONS We present the first brain autopsy findings in a molecularly proven case of ADCY5-dyskinesia, showing increased ADCY5 immunoreactivity in neurons and evidence of tau deposition. Additional patients will need to be studied to determine whether increased immunoreactivity for ADCY5 is a signature for ADCY5-dyskinesia and whether this disease has a tauopathy component.
Collapse
Affiliation(s)
- Dong‐Hui Chen
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| | - Caitlin S. Latimer
- Department of Pathology, Neuropathology DivisionUniversity of WashingtonSeattleWashingtonUSA
| | - Min Spencer
- Department of Medicine, Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
| | - Prasanthi Karna
- Department of Medicine, Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
| | - Luis F. Gonzalez‐Cuyar
- Department of Pathology, Neuropathology DivisionUniversity of WashingtonSeattleWashingtonUSA
| | - Marie Y. Davis
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
- Department of NeurologyVA Puget Sound Health Care SystemSeattleWashingtonUSA
| | - C. Dirk Keene
- Department of Pathology, Neuropathology DivisionUniversity of WashingtonSeattleWashingtonUSA
| | - Thomas D. Bird
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
- Department of Medicine, Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
- Geriatric Research, Education, and Clinical Center (GRECC)VA Puget Sound Health Care SystemSeattleWashingtonUSA
| | - Wendy H. Raskind
- Department of Medicine, Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
- Geriatric Research, Education, and Clinical Center (GRECC)VA Puget Sound Health Care SystemSeattleWashingtonUSA
- Mental Illness Research, Education, and Clinical Center (MIRECC), VA Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Psychiatry and Behavioral SciencesUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
28
|
Vijiaratnam N, Bhatia KP, Lang AE, Raskind WH, Espay AJ. ADCY5-Related Dyskinesia: Improving Clinical Detection of an Evolving Disorder. Mov Disord Clin Pract 2019; 6:512-520. [PMID: 31538084 PMCID: PMC6749814 DOI: 10.1002/mdc3.12816] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The phenotypic spectrum of adenylyl cyclase 5 (ADCY5)-related disease has expanded considerably since the first description of the disorder in 1978 as familial essential chorea in a multiplex family. OBJECTIVE To examine recent advances in the understanding of ADCY5-related dyskinesia and outline a diagnostic approach to enhance clinical detection. METHODS A pragmatic review of the ADCY5 literature was undertaken to examine unique genetic and pathophysiological features as well as distinguishing clinical features. RESULTS With over 70 cases reported to date, the phenotype is recognized to be broad, although distinctive features include prominent facial dyskinesia, motor exacerbations during drowsiness or sleep arousal, episodic painful dystonic posturing increased with stress or illness, and axial hypotonia with delayed developmental milestones. Uncommon phenotypes include childhood-onset chorea, myoclonus-dystonia, isolated nongeneralized dystonia, and alternating hemiplegia. CONCLUSION The ongoing expansion in clinical features suggests that ADCY5 remains underdiagnosed and may account for a proportion of "idiopathic" hyperkinetic movement disorders. Enhanced understanding of its clinical features may help clinicians improve the detection of complex or uncommon cases.
Collapse
Affiliation(s)
| | - Kailash P. Bhatia
- Department of Clinical and Movement Neurosciences, Queen Square Institute of NeurologyUniversity College LondonLondonUnited Kingdom
| | - Anthony E. Lang
- Department of Medicine, Division of Neurology, Edmond J. Safra Program in Parkinson's Disease, Toronto Western HospitalUniversity of TorontoTorontoOntarioCanada
| | - Wendy H. Raskind
- Departments of Medicine and Psychiatry and Behavioral SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Alberto J. Espay
- Department of Neurology (J.S.), Kingston General Hospital, Canada; Department of Neurology (D.M.‐G.), Hospital Universitario Virgen del Rocío, Seville, Spain; and UC Gardner Neuroscience Institute and Gardner Family Center for Parkinson's Disease and Movement Disorders (A.Z., A.J.E.), Department of NeurologyUniversity of CincinnatiOhioUSA
| |
Collapse
|