1
|
Dong Z, Wang Q, Yan Y, Qiang LO, Liu M. Evolution and functional divergence of the Fidgetin family. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1872:119870. [PMID: 39481482 DOI: 10.1016/j.bbamcr.2024.119870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
The Fidgetin (FIGN) family, which comprises FIGN, Fidgetin-like 1 (FIGNL1), and Fidgetin-like 2 (FIGNL2), is a vital group of microtubule-severing proteins. These proteins feature a conserved AAA+ domain essential for ATPase activity and a hexameric assembly. This review provides an in-depth analysis of the evolution and functional divergence of the FIGN family members, highlighting their role in the dynamic organization of the cytoskeleton. We further explore their broader biological functions across various species, systems, and subcellular localization. Although the FIGN family is conserved, each member exhibits unique structural characteristics and functions that reflect their evolutionary adaptations. FIGNL1 is found across animal species, while FIGNL2 is specific to vertebrates, thereby indicating its more recent evolutionary origin. Moreover, synteny analysis has revealed that FIGN is located in a more conserved genomic region compared to FIGNL2, which has undergone substantial evolutionary changes. The expression patterns of the FIGN members also vary across organisms and tissues. For example, FIGNL2 shows a notably reduced expression in the mammalian nervous system compared to that in lower vertebrates. The FIGN family members have distinct roles in microtubule severing, cell division, and DNA repair. Specifically, FIGN is involved in cell division and neuronal regeneration, FIGNL1 in axonal growth and DNA repair, and FIGNL2 in cell migration and vascular development. Their involvement in these processes underscores their role as potential biomarkers for certain cancers as well as therapeutic targets for diseases affecting the nervous system and cardiovascular development. All these evolutionary insights and functional distinctions of the FIGN family offer a comprehensive framework for understanding cytoskeletal regulation and its implications in health and disease.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Qing Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yingying Yan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Liang Oscar Qiang
- Department of Neurobiology & Anatomy at Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
2
|
Candidate Genes Encoding Dopamine Receptors as Predictors of the Risk of Antipsychotic-Induced Parkinsonism and Tardive Dyskinesia in Schizophrenic Patients. Biomedicines 2021; 9:biomedicines9080879. [PMID: 34440083 PMCID: PMC8389582 DOI: 10.3390/biomedicines9080879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/01/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Introduction: Extrapyramidal disorders form the so-called extrapyramidal syndrome (EPS), which is characterized by the occurrence of motor disorders as a result of damage to the basal ganglia and the subcortical-thalamic connections. Often, this syndrome develops while taking medications, in particular antipsychotics (APs). (2) Purpose: To review studies of candidate genes encoding dopamine receptors as genetic predictors of development of AP-induced parkinsonism (AIP) and AP-induced tardive dyskinesia (AITD) in patients with schizophrenia. (3) Materials and Methods: A search was carried out for publications of PubMed, Web of Science, Springer, and e-Library databases by keywords and their combinations over the last 10 years. In addition, the review includes earlier publications of historical interest. Despite extensive searches of these commonly used databases and search terms, it cannot be ruled out that some publications were possibly missed. (4) Results: The review considers candidate genes encoding dopamine receptors involved in pharmacodynamics, including genes DRD1, DRD2, DRD3, and DRD4. We analyzed 18 genome-wide studies examining 37 genetic variations, including single nucleotide variants (SNVs)/polymorphisms of four candidate genes involved in the development of AIP and AITD in patients with schizophrenia. Among such a set of obtained results, only 14 positive associations were revealed: rs1799732 (141CIns/Del), rs1800497 (C/T), rs6275 (C/T), rs6275 (C/T) DRD2; rs167771 (G/A) DRD3 with AIP and rs4532 (A/G) DRD1, rs6277 (C/T), rs6275 (C/T), rs1800497 (C/T), rs1079597 (A/G), rs1799732 (141CIns/Del), rs1045280 (C/G) DRD2, rs6280 (C/T), rs905568 (C/G) DRD3 with AITD. However, at present, it should be recognized that there is no final or unique decision on the leading role of any particular SNVs/polymorphisms in the development of AIP and AITD. (5) Conclusion: Disclosure of genetic predictors of the development of AIP and AITD, as the most common neurological adverse drug reactions (ADRs) in the treatment of patients with psychiatric disorders, may provide a key to the development of a strategy for personalized prevention and treatment of the considered complication of AP therapy for schizophrenia in real clinical practice.
Collapse
|
3
|
Genetic Testing for Antipsychotic Pharmacotherapy: Bench to Bedside. Behav Sci (Basel) 2021; 11:bs11070097. [PMID: 34209185 PMCID: PMC8301006 DOI: 10.3390/bs11070097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/12/2021] [Accepted: 06/23/2021] [Indexed: 11/24/2022] Open
Abstract
There is growing research interest in learning the genetic basis of response and adverse effects with psychotropic medications, including antipsychotic drugs. However, the clinical utility of information from genetic studies is compromised by their controversial results, primarily due to relatively small effect and sample sizes. Clinical, demographic, and environmental differences in patient cohorts further explain the lack of consistent results from these genetic studies. Furthermore, the availability of psychopharmacological expertise in interpreting clinically meaningful results from genetic assays has been a challenge, one that often results in suboptimal use of genetic testing in clinical practice. These limitations explain the difficulties in the translation of psychopharmacological research in pharmacogenetics and pharmacogenomics from bench to bedside to manage increasingly treatment-refractory psychiatric disorders, especially schizophrenia. Although these shortcomings question the utility of genetic testing in the general population, the commercially available genetic assays are being increasingly utilized to optimize the effectiveness of psychotropic medications in the treatment-refractory patient population, including schizophrenia. In this context, patients with treatment-refractory schizophrenia are among of the most vulnerable patients to be exposed to the debilitating adverse effects from often irrational and high-dose antipsychotic polypharmacy without clinically meaningful benefits. The primary objective of this comprehensive review is to analyze and interpret replicated findings from the genetic studies to identify specific genetic biomarkers that could be utilized to enhance antipsychotic efficacy and tolerability in the treatment-refractory schizophrenia population.
Collapse
|
4
|
Cacabelos R, Carrera I, Martínez O, Alejo R, Fernández-Novoa L, Cacabelos P, Corzo L, Rodríguez S, Alcaraz M, Nebril L, Tellado I, Cacabelos N, Pego R, Naidoo V, Carril JC. Atremorine in Parkinson's disease: From dopaminergic neuroprotection to pharmacogenomics. Med Res Rev 2021; 41:2841-2886. [PMID: 34106485 DOI: 10.1002/med.21838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 02/11/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022]
Abstract
Atremorine is a novel bioproduct obtained by nondenaturing biotechnological processes from a genetic species of Vicia faba. Atremorine is a potent dopamine (DA) enhancer with powerful effects on the neuronal dopaminergic system, acting as a neuroprotective agent in Parkinson's disease (PD). Over 97% of PD patients respond to a single dose of Atremorine (5 g, p.o.) 1 h after administration. This response is gender-, time-, dose-, and genotype-dependent, with optimal doses ranging from 5 to 20 g/day, depending upon disease severity and concomitant medication. Drug-free patients show an increase in DA levels from 12.14 ± 0.34 pg/ml to 6463.21 ± 1306.90 pg/ml; and patients chronically treated with anti-PD drugs show an increase in DA levels from 1321.53 ± 389.94 pg/ml to 16,028.54 ± 4783.98 pg/ml, indicating that Atremorine potentiates the dopaminergic effects of conventional anti-PD drugs. Atremorine also influences the levels of other neurotransmitters (adrenaline, noradrenaline) and hormones which are regulated by DA (e.g., prolactin, PRL), with no effect on serotonin or histamine. The variability in Atremorine-induced DA response is highly attributable to pharmacogenetic factors. Polymorphic variants in pathogenic (SNCA, NUCKS1, ITGA8, GPNMB, GCH1, BCKDK, APOE, LRRK2, ACMSD), mechanistic (DRD2), metabolic (CYP2D6, CYP2C9, CYP2C19, CYP3A4/5, NAT2), transporter (ABCB1, SLC6A2, SLC6A3, SLC6A4) and pleiotropic genes (APOE) influence the DA response to Atremorine and its psychomotor and brain effects. Atremorine enhances DNA methylation and displays epigenetic activity via modulation of the pharmacoepigenetic network. Atremorine is a novel neuroprotective agent for dopaminergic neurons with potential prophylactic and therapeutic activity in PD.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Iván Carrera
- Department of Health Biotechnology, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Olaia Martínez
- Department of Medical Epigenetics, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | | | | | - Pablo Cacabelos
- Department of Digital Diagnosis, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Lola Corzo
- Department of Medical Biochemistry, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Susana Rodríguez
- Department of Medical Biochemistry, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Margarita Alcaraz
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Laura Nebril
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Iván Tellado
- Department of Digital Diagnosis, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Natalia Cacabelos
- Department of Medical Documentation, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Rocío Pego
- Department of Neuropsychology, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Vinogran Naidoo
- Department of Neuroscience, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| | - Juan C Carril
- Department of Genomics & Pharmacogenomics, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Spain
| |
Collapse
|
5
|
van der Burg NC, Al Hadithy AFY, van Harten PN, van Os J, Bakker PR. The genetics of drug-related movement disorders, an umbrella review of meta-analyses. Mol Psychiatry 2020; 25:2237-2250. [PMID: 32020047 DOI: 10.1038/s41380-020-0660-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/10/2019] [Accepted: 01/17/2020] [Indexed: 12/17/2022]
Abstract
This umbrella review investigates which genetic factors are associated with drug-related movement disorders (DRMD), in an attempt to provide a synthesis of published evidence of candidate-gene studies. To identify all relevant meta-analyses, a literature search was performed. Titles and abstracts were screened by two authors and the methodological quality of included meta-analyses was assessed using 'the assessment of multiple systematic reviews' (AMSTAR) critical appraisal checklist. The search yielded 15 meta-analytic studies reporting on genetic variations in 10 genes. DRD3, DRD2, CYP2D6, HTR2A, COMT, HSPG2 and SOD2 genes have variants that may increase the odds of TD. However, these findings do not concur with early genome-wide association studies. Low-power samples are susceptible to 'winner's curse', which was supported by diminishing meta-analytic effects of several genetic variants over time. Furthermore, analyses pertaining to the same genetic variant were difficult to compare due to differences in patient populations, methods used and the choice of studies included in meta-analyses. In conclusion, DRMD is a complex phenotype with multiple genes that impact the probability of onset. More studies with larger samples using other methods than by candidate genes, are essential to developing methods that may predict the probability of DRMD. To achieve this, multiple research groups need to collaborate and a DRMD genetic database needs to be established in order to overcome winner's curse and publication bias, and to allow for stratification by patient characteristics. These endeavours may help the development of a test with clinical value in the prevention and treatment of DRMD.
Collapse
Affiliation(s)
- Nadine C van der Burg
- Zon & Schild, GGZ Centraal, Amersfoort, The Netherlands.
- Department of Psychiatry, Amsterdam UMC, Amsterdam, Netherlands.
| | | | - Peter N van Harten
- Zon & Schild, GGZ Centraal, Amersfoort, The Netherlands
- Department of Psychiatry and Psychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jim van Os
- Department of Psychiatry and Psychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre, Maastricht, The Netherlands
- Department Psychiatry, Brain Centre Rudolf Magnus, Utrecht University Medical Centre, Utrecht, The Netherlands
- Department of Psychosis Studies, King's College London, King's Health Partners, Institute of Psychiatry, London, UK
| | - P Roberto Bakker
- Zon & Schild, GGZ Centraal, Amersfoort, The Netherlands
- Department of Psychiatry and Psychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre, Maastricht, The Netherlands
- Department Psychiatry, Brain Centre Rudolf Magnus, Utrecht University Medical Centre, Utrecht, The Netherlands
| |
Collapse
|
6
|
Boloc D, Rodríguez N, Torres T, García-Cerro S, Parellada M, Saiz-Ruiz J, Cuesta MJ, Bernardo M, Gassó P, Lafuente A, Mas S, Arnaiz JA. Identifying key transcription factors for pharmacogenetic studies of antipsychotics induced extrapyramidal symptoms. Psychopharmacology (Berl) 2020; 237:2151-2159. [PMID: 32382784 DOI: 10.1007/s00213-020-05526-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We explore the transcription factors involved in the molecular mechanism of antipsychotic (AP)-induced acute extrapyramidalsymptoms (EPS) in order to identify new candidate genes for pharmacogenetic studies. METHODS Protein-protein interaction (PPI) networks previously created from three pharmacogenomic models (in vitro, animal, and peripheral blood inhumans) were used to, by means of several bioinformatic tools; identify key transcription factors (TFs) that regulate each network. Once the TFs wereidentified, SNPs disrupting the binding sites (TFBS) of these TFs in the genes of each network were selected for genotyping. Finally, SNP-basedassociations with EPS were analyzed in a sample of 356 psychiatric patients receiving AP. RESULTS Our analysis identified 33 TFs expressed in the striatum, and 125 SNPs disrupting TFBS in 50 genes of our initial networks. Two SNPs (rs938112,rs2987902) in two genes (LSMAP and ABL1) were significantly associated with AP induced EPS (p < 0.001). These SNPs disrupt TFBS regulated byPOU2F1. CONCLUSION Our results highlight the possible role of the disruption of TFBS by SNPs in the pharmacological response to AP.
Collapse
Affiliation(s)
- Daniel Boloc
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Teresa Torres
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Susana García-Cerro
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Mara Parellada
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
| | - Jeronimo Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Hospital Ramon y Cajal, Universidad de Alcala, IRYCIS, Madrid, Spain
| | - Manuel J Cuesta
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Department of Psychiatry, Complejo Hospitalario de Navarra. Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Miquel Bernardo
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Patricia Gassó
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Amalia Lafuente
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sergi Mas
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, Madrid, Spain.
- Spain The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| | - Joan Albert Arnaiz
- Dept. Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
Squassina A, Meloni A, Chillotti C, Pisanu C. Zinc finger proteins in psychiatric disorders and response to psychotropic medications. Psychiatr Genet 2019; 29:132-141. [PMID: 31464994 DOI: 10.1097/ypg.0000000000000231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Zinc finger proteins are a large family of abundantly expressed small motifs that play a crucial role in a wide range of physiological and pathophysiological mechanisms. Findings published so far support an involvement of zinc fingers in psychiatric disorders. Most of the evidence has been provided for the zinc finger protein 804A (ZNF804A) gene, which has been suggested to be implicated in schizophrenia and bipolar disorder. This evidence has been corroborated by a wide range of functional studies showing that ZNF804A regulates the expression of genes involved in cell adhesion and plays a crucial role in neurite formation and maintenance of dendritic spines. On the other hand, far less is known on other zinc finger proteins and their involvement in psychiatric disorders. In this review, we discussed studies exploring the role of zinc finger proteins in schizophrenia, bipolar disorder, and major depressive disorder as well as in pharmacogenetics of psychotropic drugs.
Collapse
Affiliation(s)
- Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari Unit of Clinical Pharmacology, University Hospital of Cagliari, Cagliari, Italy Department of Psychiatry, Dalhousie University, Halifax, NS, Canada Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
8
|
Pieters LE, Bakker PR, van Harten PN. Asymmetric Drug-Induced Parkinsonism and Psychopathology: A Prospective Naturalistic Study in Long-Stay Psychiatric Patients. Front Psychiatry 2018; 9:18. [PMID: 29459835 PMCID: PMC5807329 DOI: 10.3389/fpsyt.2018.00018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/18/2018] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Drug-induced parkinsonism (DIP) is the most common movement disorder induced by antipsychotics. Although DIP is mostly symmetric, asymmetric DIP is reported in a substantial part of the patients. We investigated the frequency of motor asymmetry in DIP and its relationship to the severity of psychopathology in long-stay psychiatric patients. METHODS We obtained data from a cohort study of 207 long-stay psychiatric patients on the frequency and risk factors of tardive dyskinesia, akathisia, tardive dystonia, and DIP. From July 2003 to May 2007 (mean follow-up, 1.1 year) drug-induced movement disorders were assessed at least two times in each patient, with a frequency of persistent DIP of 56.2%. All patients who had at least one time parkinsonism in the upper/lower limb(s) were included for analyses (190 patients, 79 women; mean age, 48.0 ± 12.9 years). The Unified Parkinson Disease Rating Scale motor scale was used to calculate the frequency of asymmetric parkinsonism. Multilevel mixed models were built to explore the relationship between asymmetry in parkinsonism and the severity of psychopathology, measured on the Clinical Global Impression-Schizophrenia scale severity index (CGI-SCH SI). RESULTS The frequency of asymmetric parkinsonism was 20.8%. Asymmetry in parkinsonism was associated with symptom severity on all CGI-SCH SI scales (β range, 0.37-3.74) and significantly associated with the positive symptom scale (β, 3.74; 95% CI, 0.35-7.31). CONCLUSION DIP is asymmetric in a substantial part of patients. Asymmetric presentation of DIP is of clinical relevance as it is related to the severity of psychopathology and may alert the clinician of more severe psychopathology. Future research is recommended to provide insight into the neuropsychopathology and clinical value of asymmetric parkinsonism for psychiatric patients.
Collapse
Affiliation(s)
- Lydia E Pieters
- Faculty of Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - P Roberto Bakker
- Psychiatric Center GGz Centraal, Amersfoort, Netherlands.,Department of Psychiatry and Psychology, Maastricht University Medical Center, South Limburg Mental Health and Teaching Network, Maastricht, Netherlands
| | - Peter N van Harten
- Psychiatric Center GGz Centraal, Amersfoort, Netherlands.,Department of Psychiatry and Psychology, Maastricht University Medical Center, South Limburg Mental Health and Teaching Network, Maastricht, Netherlands
| |
Collapse
|
9
|
Santoro ML, Moretti PN, Pellegrino R, Gadelha A, Abílio VC, Hayashi MAF, Belangero SI, Hakonarson H. A current snapshot of common genomic variants contribution in psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2016; 171:997-1005. [PMID: 27486013 DOI: 10.1002/ajmg.b.32475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/06/2016] [Indexed: 12/22/2022]
Abstract
In the past decade, numerous advances were achieved in psychiatric genetics. Particularly, the genome wide association studies (GWAS) have contributed to uncovering new genes and pathways associated to psychiatric disorders (PDs). At the same time, with increasing sample sizes in the GWAS, the polygenic risk score (PRS) promoted an additional tool for identification and evaluation the genetic risk quantitatively in PDs. This concept review presents the state of the art GWAS analysis and PRS focusing on the genetic underpinnings of PDs. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marcos L Santoro
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Genetics Division, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Patricia N Moretti
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Genetics Division, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Renata Pellegrino
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ary Gadelha
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Vanessa C Abílio
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Pharmacology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Mirian A F Hayashi
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Pharmacology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Sintia I Belangero
- Interdisciplinary Laboratory of Clinical Neurosciences, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Department of Psychiatry, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
- Genetics Division, Federal University of Sao Paulo (UNIFESP), Sao Paulo, SP, Brazil
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Mas S, Gassó P, Lafuente A, Bioque M, Lobo A, Gonzàlez-Pinto A, Olmeda MS, Corripio I, Llerena A, Cabrera B, Saiz-Ruiz J, Bernardo M. Pharmacogenetic study of antipsychotic induced acute extrapyramidal symptoms in a first episode psychosis cohort: role of dopamine, serotonin and glutamate candidate genes. THE PHARMACOGENOMICS JOURNAL 2016; 16:439-45. [PMID: 27272046 DOI: 10.1038/tpj.2016.44] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/22/2016] [Accepted: 05/02/2016] [Indexed: 01/16/2023]
Abstract
This study investigated whether the risk of presenting antipsychotic (AP)-induced extrapyramidal symptoms (EPS) could be related to single-nucleotide polymorphisms (SNPs) in a naturalistic cohort of first episode psychosis (FEP) patients. Two hundred and two SNPs in 31 candidate genes (involved in dopamine, serotonin and glutamate pathways) were analyzed in the present study. One hundred and thirteen FEP patients (43 presenting EPS and 70 non-presenting EPS) treated with high-potency AP (amisulpride, paliperidone, risperidone and ziprasidone) were included in the analysis. The statistical analysis was adjusted by age, gender, AP dosage, AP combinations and concomitant treatments as covariates. Four SNPs in different genes (DRD2, SLC18A2, HTR2A and GRIK3) contributed significantly to the risk of EPS after correction for multiple testing (P<1 × 10(-4)). These findings support the involvement of dopamine, serotonin and glutamate pathways in AP-induced EPS.
Collapse
Affiliation(s)
- S Mas
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
| | - P Gassó
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
| | - A Lafuente
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
| | - M Bioque
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Lobo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Department of Medicine and Psychiatry, Instituto de Investigación Sanitaria Aragón (IIS Aragón), University of Zaragoza, Zaragoza, Spain
| | - A Gonzàlez-Pinto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Department of Psychiatry, Hospital Universitario de Alava, University of the Basque Country, Leioa, Spain
| | - M S Olmeda
- Department of Psychiatry, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - I Corripio
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Department of Psychiatry, Hospital de Sant Pau, Barcelona, Spain
| | - A Llerena
- CICAB Clinical Research Centre, Extremadura University Hospital and Medical School Servicio Extremeño de Salud, Badajoz, Spain
| | - B Cabrera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - J Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Hospital Ramon y Cajal, Universidad de Alcala, IRYCIS, Madrid, Spain
| | - M Bernardo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM),Madrid, Spain
- Barcelona Clínic Schizophrenia Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
The identification of novel genetic variants associated with antipsychotic treatment response outcomes in first-episode schizophrenia patients. Pharmacogenet Genomics 2016; 26:235-42. [DOI: 10.1097/fpc.0000000000000213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Arranz MJ, Gallego C, Salazar J, Arias B. Pharmacogenetic studies of drug response in schizophrenia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1140554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
13
|
Mas S, Gassó P, Lafuente A. Applicability of gene expression and systems biology to develop pharmacogenetic predictors; antipsychotic-induced extrapyramidal symptoms as an example. Pharmacogenomics 2015; 16:1975-88. [PMID: 26556470 DOI: 10.2217/pgs.15.134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pharmacogenetics has been driven by a candidate gene approach. The disadvantage of this approach is that is limited by our current understanding of the mechanisms by which drugs act. Gene expression could help to elucidate the molecular signatures of antipsychotic treatments searching for dysregulated molecular pathways and the relationships between gene products, especially protein-protein interactions. To embrace the complexity of drug response, machine learning methods could help to identify gene-gene interactions and develop pharmacogenetic predictors of drug response. The present review summarizes the applicability of the topics presented here (gene expression, network analysis and gene-gene interactions) in pharmacogenetics. In order to achieve this, we present an example of identifying genetic predictors of extrapyramidal symptoms induced by antipsychotic.
Collapse
Affiliation(s)
- Sergi Mas
- Department of Pathological Anatomy, Pharmacology & Microbiology, University of Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Patricia Gassó
- Department of Pathological Anatomy, Pharmacology & Microbiology, University of Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Amelia Lafuente
- Department of Pathological Anatomy, Pharmacology & Microbiology, University of Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| |
Collapse
|
14
|
Erro R, Bhatia KP, Tinazzi M. Parkinsonism following neuroleptic exposure: A double-hit hypothesis? Mov Disord 2015; 30:780-5. [PMID: 25801826 DOI: 10.1002/mds.26209] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 01/19/2015] [Accepted: 02/06/2015] [Indexed: 01/07/2023] Open
Abstract
Drug-induced parkinsonism is caused by an offending drug and should resolve after the causative agent has been withdrawn. However, in a number of patients, symptoms persist or may even worsen over time, suggesting the development of concomitant Parkinson's disease. The prevalence estimates of Parkinson's disease after neuroleptic exposure are unexpectedly high, suggesting a causal relationship. We critically review available literature in this regard, and some pathophysiological hypotheses that might explain such a relationship are suggested. Some patients may have an undetermined genetic susceptibility to parkinsonism. We speculate that the possible neurotoxic effect of neuroleptics exerted on a susceptible dopaminergic system would lead over the long-term to a self-fostering, progressive process. Knowledge gaps and future perspectives are discussed.
Collapse
Affiliation(s)
- Roberto Erro
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, United Kingdom.,Dipartimento di Scienze Neurologiche e del Movimento, Università di Verona, Italy
| | - Kailash P Bhatia
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, United Kingdom
| | - Michele Tinazzi
- Dipartimento di Scienze Neurologiche e del Movimento, Università di Verona, Italy
| |
Collapse
|
15
|
Greenbaum L, Lerer B. Pharmacogenetics of antipsychotic-induced movement disorders as a resource for better understanding Parkinson's disease modifier genes. Front Neurol 2015; 6:27. [PMID: 25750634 PMCID: PMC4335175 DOI: 10.3389/fneur.2015.00027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 01/30/2015] [Indexed: 12/05/2022] Open
Abstract
Antipsychotic-induced movement disorders are major side effects of antipsychotic drugs among schizophrenia patients, and include antipsychotic-induced parkinsonism (AIP) and tardive dyskinesia (TD). Substantial pharmacogenetic work has been done in this field, and several susceptibility variants have been suggested. In this paper, the genetics of antipsychotic-induced movement disorders is considered in a broader context. We hypothesize that genetic variants that are risk factors for AIP and TD may provide insights into the pathophysiology of motor symptoms in Parkinson’s disease (PD). Since loss of dopaminergic stimulation (albeit pharmacological in AIP and degenerative in PD) is shared by the two clinical entities, genes associated with susceptibility to AIP may be modifier genes that influence clinical expression of PD motor sub-phenotypes, such as age at onset, disease severity, or rate of progression. This is due to their possible functional influence on compensatory mechanisms for striatal dopamine loss. Better compensatory potential might be beneficial at the early and later stages of the PD course. AIP vulnerability variants could also be related to latent impairment in the nigrostriatal pathway, affecting its functionality, and leading to subclinical dopaminergic deficits in the striatum. Susceptibility of PD patients to early development of l-DOPA induced dyskinesia (LID) is an additional relevant sub-phenotype. LID might share a common genetic background with TD, with which it shares clinical features. Genetic risk variants may predispose to both phenotypes, exerting a pleiotropic effect. According to this hypothesis, elucidating the genetics of antipsychotic-induced movement disorders may advance our understanding of multiple aspects of PD and it clinical course, rendering this a potentially rewarding field of study.
Collapse
Affiliation(s)
- Lior Greenbaum
- Department of Neurology, Sheba Medical Center at Tel Hashomer , Ramat Gan , Israel ; The Joseph Sagol Neuroscience Center, Sheba Medical Center at Tel Hashomer , Ramat Gan , Israel
| | - Bernard Lerer
- Biological Psychiatry Laboratory, Department of Psychiatry, Hadassah - Hebrew University Medical Center , Jerusalem , Israel
| |
Collapse
|
16
|
Abstract
This review considers pharmacogenetics of the so called 'second-generation' antipsychotics. Findings for polymorphisms replicating in more than one study are emphasized and compared and contrasted with larger-scale candidate gene studies and genome-wide association study analyses. Variants in three types of genes are discussed: pharmacokinetic genes associated with drug metabolism and disposition, pharmacodynamic genes encoding drug targets, and pharmacotypic genes impacting disease presentation and subtype. Among pharmacokinetic markers, CYP2D6 metabolizer phenotype has clear clinical significance, as it impacts dosing considerations for aripiprazole, iloperidone and risperidone, and variants of the ABCB1 gene hold promise as biomarkers for dosing for olanzapine and clozapine. Among pharmacodynamic variants, the TaqIA1 allele of the DRD2 gene, the DRD3 (Ser9Gly) polymorphism, and the HTR2C -759C/T polymorphism have emerged as potential biomarkers for response and/or side effects. However, large-scale candidate gene studies and genome-wide association studies indicate that pharmacotypic genes may ultimately prove to be the richest source of biomarkers for response and side effect profiles for second-generation antipsychotics.
Collapse
Affiliation(s)
- Mark D Brennan
- Department of Biochemistry & Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
17
|
Abstract
Clinicians already face "personalized" medicine every day while experiencing the great variation in toxicities and drug efficacy among individual patients. Pharmacogenetics studies are the platform for discovering the DNA determinants of variability in drug response and tolerability. Research now focuses on the genome after its beginning with analyses of single genes. Therapeutic outcomes from several psychotropic drugs have been weakly linked to specific genetic variants without independent replication. Drug side effects show stronger associations to genetic variants, including human leukocyte antigen loci with carbamazepine-induced dermatologic outcome and MC4R with atypical antipsychotic weight gain. Clinical implementation has proven challenging, with barriers including a lack of replicable prospective evidence for clinical utility required for altering medical care. More recent studies show promising approaches for reducing these barriers to routine incorporation of pharmacogenetics data into clinical care.
Collapse
|
18
|
O'Leary OF, O'Brien FE, O'Connor RM, Cryan JF. Drugs, genes and the blues: Pharmacogenetics of the antidepressant response from mouse to man. Pharmacol Biochem Behav 2014; 123:55-76. [DOI: 10.1016/j.pbb.2013.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 10/04/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022]
|
19
|
Ozdemir V, Endrenyi L, Aynacıoğlu S, Bragazzi NL, Dandara C, Dove ES, Ferguson LR, Geraci CJ, Hafen E, Kesim BE, Kolker E, Lee EJD, Llerena A, Nacak M, Shimoda K, Someya T, Srivastava S, Tomlinson B, Vayena E, Warnich L, Yaşar U. Bernard Lerer: recipient of the 2014 inaugural Werner Kalow Responsible Innovation Prize in Global Omics and Personalized Medicine (Pacific Rim Association for Clinical Pharmacogenetics). OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:211-21. [PMID: 24649998 DOI: 10.1089/omi.2014.0029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This article announces the recipient of the 2014 inaugural Werner Kalow Responsible Innovation Prize in Global Omics and Personalized Medicine by the Pacific Rim Association for Clinical Pharmacogenetics (PRACP): Bernard Lerer, professor of psychiatry and director of the Biological Psychiatry Laboratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel. The Werner Kalow Responsible Innovation Prize is given to an exceptional interdisciplinary scholar who has made highly innovative and enduring contributions to global omics science and personalized medicine, with both vertical and horizontal (transdisciplinary) impacts. The prize is established in memory of a beloved colleague, mentor, and friend, the late Professor Werner Kalow, who cultivated the idea and practice of pharmacogenetics in modern therapeutics commencing in the 1950s. PRACP, the prize's sponsor, is one of the longest standing learned societies in the Asia-Pacific region, and was founded by Kalow and colleagues more than two decades ago in the then-emerging field of pharmacogenetics. In announcing this inaugural prize and its winner, we seek to highlight the works of prize winner, Professor Lerer. Additionally, we contextualize the significance of the prize by recalling the life and works of Professor Kalow and providing a brief socio-technical history of the rise of pharmacogenetics and personalized medicine as a veritable form of 21(st) century scientific practice. The article also fills a void in previous social science analyses of pharmacogenetics, by bringing to the fore the works of Kalow from 1995 to 2008, when he presciently noted the rise of yet another field of postgenomics inquiry--pharmacoepigenetics--that railed against genetic determinism and underscored the temporal and spatial plasticity of genetic components of drug response, with invention of the repeated drug administration (RDA) method that estimates the dynamic heritabilities of drug response. The prize goes a long way to cultivate transgenerational capacity and broader cognizance of the concept and practice of responsible innovation as an important criterion of 21(st) century omics science and personalized medicine. A new call is presently in place for the 2016 PRACP Werner Kalow prize. Nominations can be made in support of an exceptional individual interdisciplinary scholar, or alternatively, an entire research team, from any region in the world with a record of highly innovative contributions to global omics science and/or personalized medicine, in the spirit of responsible innovation. The application process is straightforward, requiring a signed, 1500-word nomination letter (by the applicant or sponsor) submitted not later than May 31, 2015.
Collapse
Affiliation(s)
- Vural Ozdemir
- 1 Pacific Rim Association for Clinical Pharmacogenetics, Associate Member Society of the International Union of Basic and Clinical Pharmacology
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Brandl EJ, Kennedy JL, Müller DJ. Pharmacogenetics of antipsychotics. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2014; 59:76-88. [PMID: 24881126 PMCID: PMC4079237 DOI: 10.1177/070674371405900203] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE During the past decades, increasing efforts have been invested in studies to unravel the influence of genetic factors on antipsychotic (AP) dosage, treatment response, and occurrence of adverse effects. These studies aimed to improve clinical care by predicting outcome of treatment with APs and thus allowing for individualized treatment strategies. We highlight most important findings obtained through both candidate gene and genome-wide association studies, including pharmacokinetic and pharmacodynamic factors. METHODS We reviewed studies on pharmacogenetics of AP response and adverse effects published on PubMed until early 2012. Owing to the high number of published studies, we focused our review on findings that have been replicated in independent studies or are supported by meta-analyses. RESULTS Most robust findings were reported for associations between polymorphisms of the cytochrome P450 system, the dopamine and the serotonin transmitter systems, and dosage, treatment response, and adverse effects, such as AP-induced weight gain or tardive dyskinesia. These associations were either detected for specific medications or for classes of APs. CONCLUSION First promising and robust results show that pharmacogenetics bear promise for a widespread use in future clinical practice. This will likely be achieved by developing algorithms that will include many genetic variants. However, further investigation is warranted to replicate and validate previous findings, as well as to identify new genetic variants involved in AP response and for replication of existing findings.
Collapse
Affiliation(s)
- Eva J Brandl
- Postdoctoral Research Fellow, Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario
| | - James L Kennedy
- Head, Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario; Director, Neuroscience Research Department, Neuroscience Department, CAMH, Toronto, Ontario; l'Anson Professor of Psychiatry and Medical Science, University of Toronto, Toronto, Ontario
| | - Daniel J Müller
- Head, Pharmacogenetics Research Clinic, Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario; Associate Professor, University of Toronto, Toronto, Ontario
| |
Collapse
|
21
|
Arranz MJ, Munro JC. Toward understanding genetic risk for differential antipsychotic response in individuals with schizophrenia. Expert Rev Clin Pharmacol 2014; 4:389-405. [DOI: 10.1586/ecp.11.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Abstract
OBJECTIVE As genotyping technology has progressed, genome-wide association studies (GWAS) have matured into efficient and effective tools for mapping genes underlying human phenotypes. METHODS Recent studies have shown the utility of the GWAS approach for examining pharmacogenomic traits, including drug metabolism, efficacy, and toxicity. RESULTS Application of GWAS to pharmacogenomic outcomes presents unique challenges and opportunities. CONCLUSION In the current review, we discuss the potential promises and potential caveats of this approach specifically as it relates to pharmacogenomic studies. Concerns with study design, power and sample size, and analysis are reviewed. We further examine the features of successful pharmacogenomic GWAS, and describe consortia efforts that are likely to expand the reach of pharmacogenomic GWAS in the future.
Collapse
|
23
|
Tsermpini EE, Assimakopoulos K, Bartsakoulia M, Iconomou G, Papadima EM, Mitropoulos K, Squassina A, Patrinos GP. Individualizing clozapine and risperidone treatment for schizophrenia patients. Pharmacogenomics 2014; 15:95-110. [DOI: 10.2217/pgs.13.219] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Schizophrenia is a severe disorder that significantly affects the quality of life and total functioning of patients and their caregivers. Clozapine is the first atypical antipsychotic with fewer adverse effects and established efficacy. As a rule of thumb, risperidone is one of the most reliable and effective antipsychotics for newly diagnosed and chronic schizophrenics. Pharmacogenetic studies have identified genomic variants of candidate genes that seem to be important in the way a patient responds to treatment. The recent progress made in pharmacogenomics will improve the quality of treatment, since drug doses will be tailored to the special needs of each patient. In this article, we review the available literature attempting to delineate the role of genomic variations in clozapine and risperidone response in schizophrenic patients of various ethnicities. We conclude that pharmacogenomics for these two drugs is still not ready for implementation in the clinic.
Collapse
Affiliation(s)
- Evangelia Eirini Tsermpini
- University of Patras School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece
| | | | - Marina Bartsakoulia
- University of Patras School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece
| | - Gregoris Iconomou
- University of Patras School of Medicine, Department of Psychiatry, Rion, Patras, Greece
| | - Eleni Merkouri Papadima
- University of Patras School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece
| | | | - Alessio Squassina
- University of Cagliari, Department of Biomedical Sciences, Cagliari, Sardinia, Italy
| | - George P Patrinos
- University of Patras School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece
| |
Collapse
|
24
|
Mas S, Llerena A, Saíz J, Bernardo M, Lafuente A. Strengths and weaknesses of pharmacogenetic studies of antipsychotic drugs: the potential value of the PEPs study. Pharmacogenomics 2013; 13:1773-82. [PMID: 23171340 DOI: 10.2217/pgs.12.159] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The successful application of pharmacogenetics in routine clinical practice is still a long way from becoming a reality. In order to favor the transfer of pharmacogenetic results to clinical practice, especially in psychiatry, these studies must be optimized. This article reviews the strengths and weaknesses that characterize pharmacogenetic studies in psychiatry and condition their implementation in clinical practice. We also include recommendations for improving the design of pharmacogenetic studies, which may convert their limitations into strengths and facilitate the implementation of their results into clinical practice. Finally, we discuss the potential value of naturalistic, prospective, multicenter and coordinated projects such as the 'Phenotype-genotype and environmental interaction. Application of a predictive model in first psychotic episodes' (known as the PEPs study, from the Spanish abbreviation) in pharmacogenetic studies.
Collapse
Affiliation(s)
- Sergi Mas
- Department of Anatomic Pathology, Pharmacology & Microbiology, University of Barcelona, IDIBAPS, Casanova 143, E-08036 Barcelona, Spain
| | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Antipsychotic drugs are effective in alleviating a variety of symptoms and are medication of first choice in schizophrenia. However, a substantial interindividual variability in side effects often requires a lengthy 'trial-and-error' approach until the right medication is found for the right patient. Genetic factors have long been hypothesized to be involved and identification of related gene variants could be used to predict and tailor drug treatment. RECENT FINDINGS This review highlighting the most recent genetic findings was conducted on the two most common and most well-studied side effects: antipsychotic-induced weight gain and tardive dyskinesia. SUMMARY Regarding weight gain, most promising and most consistent findings were obtained in the serotonergic system (HTR2C) and with hypothalamic leptin-melanocortin genes, in particular with one variant close to the melanocortin-4-receptor (MC4R) gene. With respect to tardive dyskinesia, most interesting findings were generally obtained in genes related to the dopaminergic system (dopamine receptors D2 and D3), and more recently with glutamatergic system genes. Overall, genetic studies have been successful in identifying strong findings, in particular for antipsychotic-induced weight gain and to some extent for tardive dyskinesia. Apart from the need for replication studies in larger and well-characterized samples, the next challenge will be to create predictive algorithms that can be used for clinical practice.
Collapse
|
26
|
Scientific challenges and implementation barriers to translation of pharmacogenomics in clinical practice. ISRN PHARMACOLOGY 2013; 2013:641089. [PMID: 23533802 PMCID: PMC3603526 DOI: 10.1155/2013/641089] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 02/04/2013] [Indexed: 12/20/2022]
Abstract
The mapping of the human genome and subsequent advancements in genetic technology had provided clinicians and scientists an understanding of the genetic basis of altered drug pharmacokinetics and pharmacodynamics, as well as some examples of applying genomic data in clinical practice. This has raised the public expectation that predicting patients' responses to drug therapy is now possible in every therapeutic area, and personalized drug therapy would come sooner than later. However, debate continues among most stakeholders involved in drug development and clinical decision-making on whether pharmacogenomic biomarkers should be used in patient assessment, as well as when and in whom to use the biomarker-based diagnostic tests. Currently, most would agree that achieving the goal of personalized therapy remains years, if not decades, away. Realistic application of genomic findings and technologies in clinical practice and drug development require addressing multiple logistics and challenges that go beyond discovery of gene variants and/or completion of prospective controlled clinical trials. The goal of personalized medicine can only be achieved when all stakeholders in the field work together, with willingness to accept occasional paradigm change in their current approach.
Collapse
|
27
|
Antipsychotic-induced movement disorders in long-stay psychiatric patients and 45 tag SNPs in 7 candidate genes: a prospective study. PLoS One 2012; 7:e50970. [PMID: 23226551 PMCID: PMC3514178 DOI: 10.1371/journal.pone.0050970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/29/2012] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Four types of antipsychotic-induced movement disorders: tardive dyskinesia (TD), parkinsonism, akathisia and tardive dystonia, subtypes of TD (orofacial and limb truncal dyskinesia), subtypes of parkinsonism (rest tremor, rigidity, and bradykinesia), as well as a principal-factor of the movement disorders and their subtypes, were examined for association with variation in 7 candidate genes (GRIN2B, GRIN2A, HSPG2, DRD3, DRD4, HTR2C, and NQO1). METHODS Naturalistic study of 168 white long-stay patients with chronic mental illness requiring long-term antipsychotic treatment, examined by the same rater at least two times over a 4-year period, with a mean follow-up time of 1.1 years, with validated scales for TD, parkinsonism, akathisia, and tardive dystonia. The authors genotyped 45 tag SNPs in 7 candidate genes, associated with movement disorders or schizophrenia in previous studies. Genotype and allele frequency comparisons were performed with multiple regression methods for continuous movement disorders. RESULTS Various tag SNPs reached nominal significance; TD with rs1345423, rs7192557, rs1650420, as well as rs11644461; orofacial dyskinesia with rs7192557, rs1650420, as well as rs4911871; limb truncal dyskinesia with rs1345423, rs7192557, rs1650420, as well as rs11866328; bradykinesia with rs2192970; akathisia with rs324035; and the principal-factor with rs10772715. After controlling for multiple testing, no significant results remained. CONCLUSIONS The findings suggest that selected tag SNPs are not associated with a susceptibility to movement disorders. However, as the sample size was small and previous studies show inconsistent results, definite conclusions cannot be made. Replication is needed in larger study samples, preferably in longitudinal studies which take the fluctuating course of movement disorders and gene-environment interactions into account.
Collapse
|
28
|
Ni X, Zhang W, Huang RS. Pharmacogenomics discovery and implementation in genome-wide association studies era. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2012. [PMID: 23188748 DOI: 10.1002/wsbm.1199] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Clinical response to therapeutic treatments often varies among individual patients, ranging from beneficial effect to even fatal adverse reaction. Pharmacogenomics holds the promise of personalized medicine through elucidating genetic determinants responsible for pharmacological outcomes (e.g., cytotoxicities to anticancer drugs) and therefore guide the prescription decision prior to drug treatment. Besides traditional candidate gene-based approaches, technical advances have begun to allow application of whole-genome approaches to pharmacogenomic discovery. In particular, comprehensive understanding of human genetic variation provides the basis for applying GWAS (genome-wide association studies) in pharmacogenomic research to identify genomic loci associated with pharmacological phenotypes (e.g., individual dose requirement for warfarin). We therefore briefly reviewed the background for pharmacogenetic/pharmacogenomic research with statins and warfarin as examples for the GWAS discovery and their clinical implementation. In conclusion, with some challenges, whole-genome approaches such as GWAS have allowed unprecedented progress in identifying genetic variants associated with pharmacological phenotypes, as well as provided foundation for the next wave of pharmacogenomic discovery utilizing sequencing-based approaches. Furthermore, investigation of the complex interactions among genetic and epigenetic factors on the whole-genome scale will become the post-GWAS research focus for pharmacologic complex traits.
Collapse
Affiliation(s)
- Xiuqin Ni
- Department of Anatomy, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, China
| | | | | |
Collapse
|
29
|
Drago A, Giegling I, Schäfer M, Hartmann AM, Möller HJ, De Ronchi D, Stassen HH, Serretti A, Rujescu D. No association of a set of candidate genes on haloperidol side effects. PLoS One 2012; 7:e44853. [PMID: 23077486 PMCID: PMC3471928 DOI: 10.1371/journal.pone.0044853] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/08/2012] [Indexed: 11/19/2022] Open
Abstract
We previously investigated a sample of patients during an active phase of psychosis in the search for genetic predictors of haloperidol induced side effects. In the present work we extend the genetic association analysis to a wider panel of genetic variations, including 508 variations located in 96 genes. The original sample included 96 patients. An independent group of 357 patients from the CATIE study served as a replication sample. Outcomes in the investigation sample were the variation through time of: 1) the ESRS and UKU total scores 2) ESRS and UKU subscales (neurologic and psychic were included) related to tremors and 3) ESRS and UKU subscales that do not relate to tremors. Outcome in the replication sample was the presence vs absence of motoric side effects from baseline to visit 1 (∼ one month of treatment) as assessed by the AIMS scale test. Rs2242480 located in the CYP3A4 was associated with a different distribution of the UKU neurologic scores through time (permutated p = 0.047) along with a trend for a different haloperidol plasma levels (lower in CC subjects). This finding was not replicated in the CATIE sample. In conclusion, we did not find conclusive evidence for a major association between the investigated variations and haloperidol induced motoric side effects
Collapse
Affiliation(s)
- Antonio Drago
- Institute of Psychiatry, University of Bologna, Bologna, Italy
| | - Ina Giegling
- Department of Psychiatry, Ludwig Maximilians University, Munich, Germany
| | - Martin Schäfer
- Department of Psychiatry, Ludwig Maximilians University, Munich, Germany
| | | | - Hans-Jürgen Möller
- Department of Psychiatry, Ludwig Maximilians University, Munich, Germany
| | - Diana De Ronchi
- Institute of Psychiatry, University of Bologna, Bologna, Italy
| | - Hans H. Stassen
- Psychiatric University Hospital, Zurich, Zurich, Switzerland
| | | | - Dan Rujescu
- Department of Psychiatry, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
30
|
Abstract
Pharmacogenetic/pharmacogenomic (PGx) approaches to psychopharmacology aim to identify clinically meaningful predictors of drug efficacy and/or side-effect burden. To date, however, PGx studies in psychiatry have not yielded compelling results, and clinical utilization of PGx testing in psychiatry is extremely limited. In this review, the authors provide a brief overview on the status of PGx studies in psychiatry, review the commercialization process for PGx tests and then discuss methodological considerations that may enhance the potential for clinically applicable PGx tests in psychiatry. The authors focus on design considerations that include increased ascertainment of subjects in the earliest phases of illness, discuss the advantages of drug-induced adverse events as phenotypes for examination and emphasize the importance of maximizing adherence to treatment in pharmacogenetic studies. Finally, the authors discuss unique aspects of pharmacogenetic studies that may distinguish them from studies of other complex traits. Taken together, these data provide insights into the design and methodological considerations that may enhance the potential for clinical utility of PGx studies.
Collapse
|
31
|
Bakker PR, Bakker E, Amin N, van Duijn CM, van Os J, van Harten PN. Candidate gene-based association study of antipsychotic-induced movement disorders in long-stay psychiatric patients: a prospective study. PLoS One 2012; 7:e36561. [PMID: 22615781 PMCID: PMC3352907 DOI: 10.1371/journal.pone.0036561] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 04/05/2012] [Indexed: 12/20/2022] Open
Abstract
Objective Four types of antipsychotic-induced movement disorders: tardive dyskinesia (TD), parkinsonism, akathisia and tardive dystonia, subtypes of TD (orofacial and limb truncal dyskinesia), subtypes of parkinsonism (rest tremor, rigidity, and bradykinesia), as well as a principal-factor of the movement disorders and their subtypes, were examined for association with variation in 10 candidate genes (PPP1R1B, BDNF, DRD3, DRD2, HTR2A, HTR2C, COMT, MnSOD, CYP1A2, and RGS2). Methods Naturalistic study of 168 white long-stay patients with chronic mental illness requiring long-term antipsychotic treatment, examined by the same rater at least two times over a 4-year period, with a mean follow-up time of 1.1 years, with validated scales for TD, parkinsonism, akathisia, and tardive dystonia. The authors genotyped 31 SNPs, associated with movement disorders or schizophrenia in previous studies. Genotype and allele frequency comparisons were performed with multiple regression methods for continuous movement disorders. Results Various SNPs reached nominal significance: TD and orofacial dyskinesia with rs6265 and rs988748, limb truncal dyskinesia with rs6314, rest tremor with rs6275, rigidity with rs6265 and rs4680, bradykinesia with rs4795390, akathisia with rs4680, tardive dystonia with rs1799732, rs4880 and rs1152746. After controlling for multiple testing, no significant results remained. Conclusions The findings suggest that selected SNPs are not associated with a susceptibility to movement disorders. However, as the sample size was small and previous studies show inconsistent results, definite conclusions cannot be made. Replication is needed in larger study samples, preferably in longitudinal studies which take the fluctuating course of movement disorders and gene-environment interactions into account.
Collapse
|
32
|
Association of the ZFPM2 gene with antipsychotic-induced parkinsonism in schizophrenia patients. Psychopharmacology (Berl) 2012; 220:519-28. [PMID: 21947317 DOI: 10.1007/s00213-011-2499-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/09/2011] [Indexed: 02/06/2023]
Abstract
RATIONALE Antipsychotic-induced parkinsonism (AIP) is a severe adverse affect of antipsychotic drug treatment. Recently, our group performed a genome-wide association study (GWAS) for AIP severity, and identified several potential AIP risk variants. OBJECTIVES The aim of this study was to validate our original AIP-GWAS susceptibility variants and to understand their possible function. METHODS We conducted a validation study of 15 single-nucleotide polymorphisms (SNPs) in an independent sample of 178 US schizophrenia patients treated for at least a month with typical or atypical antipsychotics. Then, a sample of 49 Jewish Israeli Parkinson's disease (PD) patients with available neuroimaging ([(123)I]-FP-CIT-SPECT) data was analyzed, to study association of confirmed AIP SNPs with level of dopaminergic deficits in the putamen. RESULTS Using logistic regression and controlling for possible confounders, we found nominal association of the intronic SNP, rs12678719, in the Zinc Finger Protein Multitype 2 (ZFPM2) gene with AIP (62 affected/116 unaffected), in the whole sample (p = 0.009; P = 5.97 × 10(-5) in the GWAS), and in the African American sub-sample (N = 111; p = 0.002). The same rs12678719-G AIP susceptibility allele was associated with lower levels of dopaminergic neuron related ligand binding in the contralateral putamen of PD patients (p = 0.026). CONCLUSIONS Our preliminary findings support association of the ZFPM2 SNP, rs12678719, with AIP. At the functional level, this variant is associated with deficits in the nigrostriatal pathway in PD patients that may be related to latent subclinical deficits among AIP-prone individuals with schizophrenia. Further validation studies in additional populations are required.
Collapse
|
33
|
Kasarskis A, Yang X, Schadt E. Integrative genomics strategies to elucidate the complexity of drug response. Pharmacogenomics 2012; 12:1695-715. [PMID: 22118053 DOI: 10.2217/pgs.11.115] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pharmacogenomic investigation from both genome-wide association studies and experiments focused on candidate loci involved in drug mechanism and metabolism has yielded a substantial and increasing list of robust genetic effects on drug therapy in humans. At the same time, reasonably comprehensive molecular data such as gene expression, proteomic and metabolomic data are now available for collections of hundreds to thousands of individuals. If these data are structured in a statistically robust and computationally tractable way, such as a network model, they can aid in the analysis of new pharmacogenomics studies by suggesting novel hypotheses for the regulation of genes involved in drug metabolism and response. Similarly, hypotheses taken from these same models can direct genome-wide association studies by focusing the genome-wide association studies analysis on a number of specific hypotheses informed by the relationships customarily seen between a gene's expression or protein activity and genetic variation at a particular locus. Network models based on other sorts of systematic biological data such as cell-based surveys of drug effect on gene expression and mining of literature and electronic medical records for associations between clinical and molecular phenotypes also promise similar utility. Although surely primitive in comparison with what will be developed, these model-based approaches to leveraging the increasing volume of data generated in the course of patient care and medical research nevertheless suggest a huge opportunity to improve our understanding of biological systems involved in pharmacogenomics and apply them to questions of medical relevance.
Collapse
|
34
|
|
35
|
Koning JP, Vehof J, Burger H, Wilffert B, Al Hadithy A, Alizadeh B, van Harten PN, Snieder H. Association of two DRD2 gene polymorphisms with acute and tardive antipsychotic-induced movement disorders in young Caucasian patients. Psychopharmacology (Berl) 2012; 219:727-36. [PMID: 21750899 DOI: 10.1007/s00213-011-2394-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 06/21/2011] [Indexed: 12/13/2022]
Abstract
RATIONALE Pharmacogenetic studies on antipsychotic-induced movement disorders (MD) in schizophrenia so far have focused mainly on tardive dyskinesia. Only a few examined the more acute antipsychotic-induced MD such as parkinsonism and akathisia. Notably, all MD relate to deregulation of the dopamine system. OBJECTIVE This study aimed to replicate previously reported associations in candidate genes for acute and tardive antipsychotic-induced MD in a young Caucasian sample. METHODS In 402 patients (median age 26 years), a total of 13 polymorphisms were genotyped in eight dopamine-related candidate genes selected a priori from the literature (regarding dopamine and serotonin receptors, dopamine degradation, and free radicals scavenging enzymes pathways). RESULTS Patients with MD used on average a higher haloperidol dose equivalent when compared to those without MD. The prevalence of MD was high and did not differ between first- and second-generation antipsychotics. Significant associations were found between (a) the TaqI_D polymorphism and akathisia (OR = 2.3, p = 0.001 for each extra C-allele) and (b) the -141C polymorphism and tardive dyskinesia (OR = 0.20, p = 0.001 for each extra Del allele). The other polymorphisms were not significantly associated with an MD. CONCLUSIONS Two associations were found between genetic variation TaqI_D and the -141C polymorphisms in the DRD2 gene and antipsychotic-induced MD; one with acute akathisia and one with tardive dyskinesia. These were previously reported to be associated with tardive dyskinesia and acute parkinsonism, respectively. These results suggest that the contribution of these DRD2 gene variants in the vulnerability of antipsychotic-induced MD takes place in a more general or pleiotropic way.
Collapse
Affiliation(s)
- Jeroen P Koning
- Mental Health Organization GGZ Centraal, Amersfoort, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
There is substantial interindividual variability in the effects of treatment with antipsychotic drugs not only in the emergence of adverse effects but also in symptom response. It is becoming increasingly clear that much of this variability is due to genetic factors; pharmacogenetics is the study of those factors, with the eventual goal of identifying genetic predictors of treatment effects. There have been many reported associations of single nucleotide polymorphisms (SNPs) in candidate genes with the consequences of antipsychotic drug treatment. Thus variations in dopaminergic and serotoninergic genes may influence positive and negative symptom outcome, respectively. Among the adverse effects, tardive dyskinesia and weight gain have been the most studied, with some consistent associations of functional SNPs in genes relating to pharmacological mechanisms. Technological advance has permitted large-scale genome-wide association studies (GWAS), but as yet there are few reports that replicate prior findings with candidate genes. Nevertheless, GWAS may identify associations which provide new clues relating to underlying mechanisms.
Collapse
Affiliation(s)
- Gavin P Reynolds
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| |
Collapse
|
37
|
Drago A, Crisafulli C, Serretti A. The genetics of antipsychotic induced tremors: a genome-wide pathway analysis on the STEP-BD SCP sample. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:975-86. [PMID: 21990027 DOI: 10.1002/ajmg.b.31245] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 09/16/2011] [Indexed: 11/08/2022]
Abstract
Extrapyramidal symptoms (EPS) are associated with antipsychotic treatment. The exact definition of the genetic variants that influence the antipsychotic induced EPS would dramatically increase the quality of antipsychotic prescriptions. We investigated this issue in a subsample of the Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD). Four hundred nine manic patients were treated with antipsychotics and had complete clinical and genetic data. Outcome was an item of the Clinical Monitoring Form which scored tremors from 0 to 4 at each clinical visit. Visits were scheduled according to clinical issues, based on a naturalistic approach. A genomic inflation factor of 1.017 resulted after genetic quality control. Single SNPs GWAS (Plink) and molecular pathway GWAS were conducted (SNP ratio test, KEGG depository). No single SNP reached GWAS significance level of association. Molecular pathways related to cell survival events and lipid synthesis were significantly associated with antipsychotic induced EPS (P = 0.0009 for Hsa04512, Hsa01031, Hsa00230, Hsa04510, Hsa03320, Hsa04930, and Hsa04115; P = 0.0019 for Hsa04020 and Hsa00561). This finding was consistent with previous GWAS studies.
Collapse
Affiliation(s)
- Antonio Drago
- Institute of Psychiatry, University of Bologna, Bologna, Italy
| | | | | |
Collapse
|
38
|
Valiente A, Lafuente A, Bernardo M. [Systematic review of the Genomewide Association Studies (GWAS) in schizophrenia]. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2011; 4:218-27. [PMID: 23446268 DOI: 10.1016/j.rpsm.2011.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 08/01/2011] [Accepted: 09/30/2011] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Heritability in schizophrenia can reach up to 80% and the risk in families is 5-10 times higher than in the general population. The large contribution of genetics in this disorder has led to a growing interest in its study. OBJECTIVES To review the findings of genetic studies known as Genomewide Association Studies (GWAS) on schizophrenia. METHOD Systematic search using Pubmed with the key words GWAS and (psychosis) or (schizophrenia). The following web pages have been reviewed: http://www.szgene.org/largescale.asp and www.genome.gov/gwastudies/. RESULTS The GWAS have focused on causal biological aspects, such as the histocompatibility complex, glutamate metabolism, apoptosis and inflammatory processes, and the immune system (TNF-β, TNFR1). Also focused in the search were the genes that modulate the appearance of secondary metabolic and cardiac effects and secondary effects in subjects with schizophrenia and on anti-psychotic treatment. In neurorecognition, over-expression of the MET proto-oncogene (MET) has been associated with a low susceptibility for schizophrenia and a better cognitive performance, as well as a lower susceptibility for the incidence of cancer. Mention is also made of the different genes that mediate in cognitive functioning depending on the anti-psychotic treatment received. CONCLUSIONS The main interests of the GWAS during the last few years have been the neurobiological pathways involved in schizophrenia. The discoveries arising from these studies have been limited. This has led to an innovative approach on the aetiological study of the disorder by studying gene-environment interactions.
Collapse
Affiliation(s)
- Alicia Valiente
- Programa Esquizofrènia Clínic, Servei de Psiquiatria, Hospital Clínic, IDIBAPS, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, España.
| | | | | |
Collapse
|
39
|
Abstract
This review presents the findings of pharmacogenetic studies exploring the influence of gene variants on antipsychotic treatment response, in terms of both symptom improvement and adverse effects, in patients with schizophrenia. Despite numerous studies in the field, replicating findings across different cohorts that include subjects of different ethnic groups has been challenging. It is clear that non-genetic factors have an important contribution to antipsychotic treatment response. Differing clinical, demographic and environmental characteristics of the cohorts studied have added substantial complexity to the interpretation of the positive and negative findings of many studies. Pharmacogenomic genome-wide investigations are beginning to yield interesting data although they have failed to replicate the most robust findings of candidate gene studies, and are limited by the sample size, especially given the need for studying homogeneous cohorts. Most of the studies conducted on cohorts treated with single antipsychotics have investigated clozapine, olanzapine or risperidone response. These studies have provided some of the most replicated associations with treatment efficacy. Serotonergic system gene variants are significantly associated with the efficacy of clozapine and risperidone, but may have less influence on the efficacy of olanzapine. Dopamine D3 receptor polymorphisms have been more strongly associated with the efficacy of clozapine and olanzapine, and D2 genetic variants with the efficacy of risperidone. Serotonin influences the control of feeding behaviour and has been hypothesized to have a role in the development of antipsychotic-induced weight gain. Numerous studies have linked the serotonin receptor 2C (5-HT2C) -759-C/T polymorphism with weight gain. The leptin gene variant, -2548-G/A, has also been associated with weight gain in several studies. Pharmacogenetic studies support the role of cytochrome P450 enzymes and dopamine receptor variants in the development of antipsychotic-induced movement disorders, with a contribution of serotonergic receptors and other gene variants implicated in the mechanism of action of antipsychotics. Clozapine-induced agranulocytosis has been associated with polymorphisms in the major histocompatibility complex gene (HLA).
Collapse
Affiliation(s)
- Maria J Arranz
- Department of Psychological Medicine, Institute of Psychiatry, Kings College London, London, UK.
| | | | | |
Collapse
|
40
|
Aragam N, Wang KS, Pan Y. Genome-wide association analysis of gender differences in major depressive disorder in the Netherlands NESDA and NTR population-based samples. J Affect Disord 2011; 133:516-21. [PMID: 21621269 DOI: 10.1016/j.jad.2011.04.054] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a universally prevalent, genetic, and environment dependent mental condition that disables people of every culture, race, gender, and age. While the gender differences for MDD have been widely reported in literature, few genome-wide analyses of gender differences have been reported to date. METHODS We conducted a genome-wide association analysis of gender differences for MDD using the Netherlands NESDA and NTR population-based samples (1726 cases and 1630 controls). PLINK software was used to analyze the genome-wide association data of Perlegen 600 K SNP Chips. RESULTS We identified 40 male-specific and 56 female-specific MDD associated SNPs with P-values less than 10(-4). The best male-specific SNP was rs9352774 (P=2.26 × 10(-6)) within LGSN gene while the best female-specific SNP was rs2715148 (P=5.64 × 10(-7)) within PCLO gene. We also found 38 SNPs showing gene × gender interactions in influencing MDD (P<10(-4)). The best SNP was rs12692709 (P=5.75 × 10(-6)) near FIGN gene at 2q24.3 while the next best SNP was rs11039588 (P=1.16 × 10(-5)) within OR4B1 gene. LIMITATIONS The findings from this study need be replicated in other populations. CONCLUSIONS These results provide genetic basis for gender differences in MDD and will serve as a resource for replication in other populations to elucidate the potential role of these genetic variants in MDD.
Collapse
Affiliation(s)
- Nagesh Aragam
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA
| | | | | |
Collapse
|
41
|
Risk for antipsychotic-induced extrapyramidal symptoms: influence of family history and genetic susceptibility. Psychopharmacology (Berl) 2011; 214:729-36. [PMID: 21072501 DOI: 10.1007/s00213-010-2079-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 10/27/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVES This study aims to further evaluate the impact of family history of primary movement disorders (FHpMD) and a candidate genetic variant on risk of antipsychotic-induced extrapyramidal symptoms (EPS). METHODS We examined 156 (76 men) inpatients receiving antipsychotics for EPS and FHpMD stratified by patient characteristics. The genetic analysis included genotyping of a multiallelic dinucleotide polymorphism in the ATP1A3 gene. RESULTS EPS lifetime prevalence was 69% and more frequent in the presence of FHpMD (p = 0.052), particularly in patients younger than 60 years (p = 0.012) and with acute dystonic reactions. The ATP1A3 polymorphism showed an allele length-dependent association with parkinsonism (p=0.019 uncorrected, p=0.057 corrected) exclusively. Carriers of the shortest allele had a 7.7-fold increased risk for parkinsonism. CONCLUSIONS The association of FHpMD and EPS may be linked to the EPS subtype and age of the patient. A common ATP1A3 genomic variation may represent a susceptibility factor for the risk for antipsychotic-induced parkinsonism in an allele-dependent manner.
Collapse
|
42
|
Lafuente A. Strategies, strengths and limitations in pharmacogenetic studies of antipsychotics. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2011; 4:69-71. [PMID: 23446141 DOI: 10.1016/j.rpsm.2011.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 06/01/2023]
|
43
|
Zhang JP, Malhotra AK. Pharmacogenetics and antipsychotics: therapeutic efficacy and side effects prediction. Expert Opin Drug Metab Toxicol 2011; 7:9-37. [PMID: 21162693 DOI: 10.1517/17425255.2011.532787] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
IMPORTANCE OF THE FIELD Antipsychotic drug is the mainstay of treatment for schizophrenia, and there are large inter-individual differences in clinical response and side effects. Pharmacogenetics provides a valuable tool to fulfill the promise of personalized medicine by tailoring treatment based on one's genetic markers. AREAS COVERED IN THIS REVIEW This article reviews the pharmacogenetic literature from early 1990s to 2010, focusing on two aspects of drug action: pharmacokinetics and pharmacodynamics. Genetic variants in the neurotransmitter receptors including dopamine and 5-HT and metabolic pathways of drugs including CYP2D6 and COMT were discussed in association with clinical drug response and side effects. WHAT THE READER WILL GAIN Readers are expected to learn the up-to-date evidence in pharmacogenetic research and to gain familiarity to the issues and challenges facing the field. TAKE HOME MESSAGE Pharmacogenetic research of antipsychotic drugs is both promising and challenging. There is consistent evidence that some genetic variants can affect clinical response and side effects. However, more studies that are designed specifically to test pharmacogenetic hypotheses are clearly needed to advance the field.
Collapse
Affiliation(s)
- Jian-Ping Zhang
- The Zucker Hillside Hospital, Feinstein Institute of Medical Research, North Shore-Long Island Jewish Health System, Division of Psychiatry Research, Department of Psychiatry, Glen Oaks, NY 11004, USA.
| | | |
Collapse
|
44
|
Abstract
Central nervous system disorders are the third greatest health problem in developed countries, and schizophrenia represents some of the most disabling ailments in young individuals. There is an abuse and/or misuse of antipsychotics, and recent advances in pharmacogenomics pose new challenges for the clinical management of this complex disorder. Schizophrenia is a multi-factorial/polygenic complex disorder in which hundreds of different genes are potentially involved, leading to the phenotypic expression of the disease in conjunction with epigenetic and environmental phenomena. Consequently, structural and functional genomic changes induce proteomic and metabolomic defects associated with the disease phenotype. Disease-related genomic profiles and genetic variants in genes involved in drug metabolism are responsible for drug efficacy and safety. About 20% of Caucasians are defective in CYP2D6 enzymes, which participate in the metabolism of 25-30% of central nervous system drugs. Approximately 40% of antipsychotics are substrates of CYP2D6 enzymes, 23% are substrates of CYP3A4, and 18% are substrates of CYP1A2. In order to achieve a mature discipline of pharmacogenomics of schizophrenia it would be effective to accelerate: (i) the education of physicians and the public in the use of genomic screening in daily clinical practice; (ii) the standardization of genetic testing for major categories of drugs; (iii) the validation of pharmacogenomic procedures according to drug category and pathology; (iv) the regulation of ethical, social, and economic issues; and (v) the incorporation of pharmacogenomic procedures of drugs in development and drugs on the market in order to optimize therapeutics.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute for CNS Disorders and Genomic Medicine, Bergondo, Coruña, Spain
| | | | | |
Collapse
|
45
|
Abstract
Existing psychotropic medications for the treatment of mental illnesses, including antidepressants, mood stabilizers, and antipsychotics, are clinically suboptimal. They are effective in only a subset of patients or produce partial responses, and they are often associated with debilitating side effects that discourage adherence. There is growing enthusiasm in the promise of pharmacogenetics to personalize the use of these treatments to maximize their efficacy and tolerability; however, there is still a long way to go before this promise becomes a reality. This article reviews the progress that has been made in research toward understanding how genetic factors influence psychotropic drug responses and the challenges that lie ahead in translating the research findings into clinical practices that yield tangible benefits for patients with mental illnesses.
Collapse
Affiliation(s)
- Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Hampton House, Room 857, 624 North Broadway, Baltimore, MD 21205, USA.
| | | |
Collapse
|
46
|
Phosphodiesterase 4B genetic variants are not associated with antipsychotic-induced tardive dyskinesia. Int Clin Psychopharmacol 2010; 25:264-9. [PMID: 20436352 DOI: 10.1097/yic.0b013e32833a5ff9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Phosphodiesterase 4B (PDE4B) has been evaluated as a genetic risk factor for schizophrenia. Selective PDE4 inhibitor drugs have antipsychotic-like effects and reduce tardive dyskinesia-like movements in animal models. We investigated whether PDE4B genetic variants are associated with antipsychotic-induced tardive dyskinesia incidence and severity in schizophrenia patients. Our sample consisted of 169 Caucasian patients taking typical antipsychotic medication for at least 1 year. We found two PDE4B gene variants to be nominally associated with tardive dyskinesia (rs1338719 and rs7528545) in the overall population and two other variants nominally associated with the presence of tardive dyskinesia and severity in female patients (rs1890196 and rs783036). None of these results survived correction for multiple testing. Overall, our results do not support a genetic association between tardive dyskinesia and PDE4B.
Collapse
|
47
|
Gurwitz D, Pirmohamed M. Pharmacogenomics: the importance of accurate phenotypes. Pharmacogenomics 2010; 11:469-70. [PMID: 20350123 DOI: 10.2217/pgs.10.41] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lack of knowledge regarding genotype-phenotype correlations is often cited as the major barrier delaying the uptake of pharmacogenomics into routine medical practice. When we look forward to genome-wide association studies as one of the most promising tools for overcoming the pharmacogenomics knowledge barrier, we must keep in mind that having large patient cohorts may not help improve our understanding of alleles implicated in drug-response phenotypes, unless we ensure that such phenotypes are precise and pertinent. It may be wiser, and far more cost effective, to invest scarce research funding in accurate patient drug-response phenotyping than to genotype (or fully sequence) hundreds to thousands of study participants. Biobanks created with personalized medicine research in mind should, when possible, have access to donors' clinical data, including detailed disease- and drug-response phenotypes.
Collapse
Affiliation(s)
- David Gurwitz
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel.
| | | |
Collapse
|
48
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2010. [DOI: 10.1002/pds.1849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Abstract
Existing psychotropic medications for the treatment of mental illnesses, including antidepressants, mood stabilizers, and antipsychotics, are clinically suboptimal. They are effective in only a subset of patients or produce partial responses, and they are often associated with debilitating side effects that discourage adherence. There is growing enthusiasm in the promise of pharmacogenetics to personalize the use of these treatments to maximize their efficacy and tolerability; however, there is still a long way to go before this promise becomes a reality. This article reviews the progress that has been made in research toward understanding how genetic factors influence psychotropic drug responses and the challenges that lie ahead in translating the research findings into clinical practices that yield tangible benefits for patients with mental illnesses.
Collapse
Affiliation(s)
- Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Hampton House, Baltimore, MD 21205, USA.
| | | |
Collapse
|
50
|
Collins LE, Galtieri DJ, Brennum LT, Sager TN, Hockemeyer J, Müller CE, Hinman JR, Chrobak JJ, Salamone JD. Oral tremor induced by the muscarinic agonist pilocarpine is suppressed by the adenosine A2A antagonists MSX-3 and SCH58261, but not the adenosine A1 antagonist DPCPX. Pharmacol Biochem Behav 2009; 94:561-9. [PMID: 19958787 DOI: 10.1016/j.pbb.2009.11.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 11/19/2009] [Accepted: 11/25/2009] [Indexed: 01/05/2023]
Abstract
Tremulous jaw movements in rats, which can be induced by dopamine (DA) antagonists, DA depletion, and cholinomimetics, have served as a useful model for studies of tremor. Although adenosine A(2A) antagonists can reduce the tremulous jaw movements induced by DA antagonists and DA depletion, there are conflicting reports about the interaction between adenosine antagonists and cholinomimetic drugs. The present studies investigated the ability of adenosine antagonists to reverse the tremorogenic effect of the muscarinic agonist pilocarpine. While the adenosine A(2A) antagonist MSX-3 was incapable of reversing the tremulous jaw movements induced by the 4.0mg/kg dose of pilocarpine, both MSX-3 and the adenosine A(2A) antagonist SCH58261 reversed the tremulous jaw movements elicited by 0.5mg/kg pilocarpine. Systemic administration of the adenosine A(1) antagonist DPCPX failed to reverse the tremulous jaw movements induced by either an acute 0.5mg/kg dose of the cholinomimetic pilocarpine or the DA D2 antagonist pimozide, indicating that the tremorolytic effects of adenosine antagonists may be receptor subtype specific. Behaviorally active doses of MSX-3 and SCH 58261 showed substantial in vivo occupancy of A(2A) receptors, but DPCPX did not. The results of these studies support the use of adenosine A(2A) antagonists for the treatment of tremor.
Collapse
Affiliation(s)
- Lyndsey E Collins
- Department of Psychology, University of Connecticut, Storrs, CT 06269-1020, USA
| | | | | | | | | | | | | | | | | |
Collapse
|