1
|
Paribello P, Isayeva U, Pisanu C, Squassina A, Manchia M. Pharmacogenomics and response to lithium in bipolar disorder. Pharmacogenomics 2025; 25:689-706. [PMID: 39998910 PMCID: PMC11901374 DOI: 10.1080/14622416.2025.2470605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
AIMS The present review explores the existing evidence on pharmacogenomic tests for prediction of lithium response in the treatment of bipolar disorder. We focused our research article on reports describing findings from genome-wide association studies, polygenic risk scores, and gene expression analyses associated with lithium response. METHODS We conducted a non-systematic review of studies from PubMed/Medline by using terms such as "pharmacogenomics," "GWAS," "gene expression," and "lithium response." Inclusion criteria focused on original research involving human subjects and assessing lithium response outcomes as well as in vitro studies. An extensive pearl-growing strategy was employed to further enlarge the scope of the piece by capitalizing on the knowledge of study authors on the subject. RESULTS The observed results, albeit promising, remain preliminary in terms of clinical relevance. Machine learning combining genetic and clinical data appears associated with moderate success in predicting lithium responsiveness. Gene expression studies and genome-wide association studies have helped identify possible targets of lithium and have the potential to support target-specific drug research. CONCLUSIONS Pharmacogenomics may support further discoveries in precision medicine further enlarging our understanding of the underlying mechanisms of lithium for its efficacy. However, clinical applications currently appear out of reach in the near future.
Collapse
Affiliation(s)
- Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Ulker Isayeva
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
2
|
Bortolozzi A, Fico G, Berk M, Solmi M, Fornaro M, Quevedo J, Zarate CA, Kessing LV, Vieta E, Carvalho AF. New Advances in the Pharmacology and Toxicology of Lithium: A Neurobiologically Oriented Overview. Pharmacol Rev 2024; 76:323-357. [PMID: 38697859 PMCID: PMC11068842 DOI: 10.1124/pharmrev.120.000007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 05/05/2024] Open
Abstract
Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Giovanna Fico
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michael Berk
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Marco Solmi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michele Fornaro
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Joao Quevedo
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Carlos A Zarate
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Lars V Kessing
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Andre F Carvalho
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| |
Collapse
|
3
|
Jitte S, Keluth S, Bisht P, Wal P, Singh S, Murti K, Kumar N. Obesity and Depression: Common Link and Possible Targets. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1425-1449. [PMID: 38747226 DOI: 10.2174/0118715273291985240430074053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/15/2024] [Accepted: 03/27/2024] [Indexed: 10/22/2024]
Abstract
Depression is among the main causes of disability, and its protracted manifestations could make it even harder to treat metabolic diseases. Obesity is linked to episodes of depression, which is closely correlated to abdominal adiposity and impaired food quality. The present review is aimed at studying possible links between obesity and depression along with targets to disrupt it. Research output in Pubmed and Scopus were referred for writing this manuscript. Obesity and depression are related, with the greater propensity of depressed people to gain weight, resulting in poor dietary decisions and a sedentary lifestyle. Adipokines, which include adiponectin, resistin, and leptin are secretory products of the adipose tissue. These adipokines are now being studied to learn more about the connection underlying obesity and depression. Ghrelin, a gut hormone, controls both obesity and depression. Additionally, elevated ghrelin levels result in anxiolytic and antidepressant-like effects. The gut microbiota influences the metabolic functionalities of a person, like caloric processing from indigestible nutritional compounds and storage in fatty tissue, that exposes an individual to obesity, and gut microorganisms might connect to the CNS through interconnecting pathways, including neurological, endocrine, and immunological signalling systems. The alteration of brain activity caused by gut bacteria has been related to depressive episodes. Monoamines, including dopamine, serotonin, and norepinephrine, have been widely believed to have a function in emotions and appetite control. Emotional signals stimulate arcuate neurons in the hypothalamus that are directly implicated in mood regulation and eating. The peptide hormone GLP-1(glucagon-like peptide- 1) seems to have a beneficial role as a medical regulator of defective neuroinflammation, neurogenesis, synaptic dysfunction, and neurotransmitter secretion discrepancy in the depressive brain. The gut microbiota might have its action in mood and cognition regulation, in addition to its traditional involvement in GI function regulation. This review addressed the concept that obesity-related low-grade mild inflammation in the brain contributes to chronic depression and cognitive impairments.
Collapse
Affiliation(s)
- Srikanth Jitte
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| | - Saritha Keluth
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| | - Priya Bisht
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| | - Pranay Wal
- PSIT- Pranveer Singh Institute of Technology, Pharmacy, Kanpur 209305, Uttar Pradesh, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali 844102, Bihar, India
| |
Collapse
|
4
|
Epigenetic signatures in antidepressant treatment response: a methylome-wide association study in the EMC trial. Transl Psychiatry 2022; 12:268. [PMID: 35794104 PMCID: PMC9259740 DOI: 10.1038/s41398-022-02032-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 12/02/2022] Open
Abstract
Although the currently available antidepressants are well established in the treatment of the major depressive disorder (MDD), there is strong variability in the response of individual patients. Reliable predictors to guide treatment decisions before or in an early stage of treatment are needed. DNA-methylation has been proven a useful biomarker in different clinical conditions, but its importance for mechanisms of antidepressant response has not yet been determined. 80 MDD patients were selected out of >500 participants from the Early Medication Change (EMC) cohort with available genetic material based on their antidepressant response after four weeks and stratified into clear responders and age- and sex-matched non-responders (N = 40, each). Early improvement after two weeks was analyzed as a secondary outcome. DNA-methylation was determined using the Illumina EPIC BeadChip. Epigenome-wide association studies were performed and differentially methylated regions (DMRs) identified using the comb-p algorithm. Enrichment was tested for hallmark gene-sets and in genome-wide association studies of depression and antidepressant response. No epigenome-wide significant differentially methylated positions were found for treatment response or early improvement. Twenty DMRs were associated with response; the strongest in an enhancer region in SORBS2, which has been related to cardiovascular diseases and type II diabetes. Another DMR was located in CYP2C18, a gene previously linked to antidepressant response. Results pointed towards differential methylation in genes associated with cardiac function, neuroticism, and depression. Linking differential methylation to antidepressant treatment response is an emerging topic and represents a step towards personalized medicine, potentially facilitating the prediction of patients' response before treatment.
Collapse
|
5
|
Genetic polymorphisms influencing response to lithium in early-onset Bipolar disorder from south India. Asian J Psychiatr 2022; 70:103018. [PMID: 35158158 DOI: 10.1016/j.ajp.2022.103018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/16/2021] [Accepted: 02/06/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Early-onset Bipolar disorder (EOBD), has a more malignant course with high recurrence risk and there is a need for population-specific pharmaco-genomic study. METHODS This study is a prospective and retrospective observational study. Both newly diagnosed patients and those on follow-up with a diagnosis of bipolar I disorder with onset before 18 years of age and on lithium prophylaxis as part of treatment-as-usual were recruited for the study. Response to treatment was assessed at the end of two years follow up using ALDA scale. Ten single nucleotide polymorphisms associated with treatment response based on previous studies were chosen for analysis. RESULTS Of 162 who had EOBD, sixty-four fulfilled inclusion criteria and fifty-seven completed the study. TT and TG genotypes of rs75222709 on AL157359.3 gene were found to be significantly different between non-responders(N = 43) and healthy controls (N = 220). The frequency of the GA genotype of the single nucleotide polymorphism rs17204573 of the RORA (Retinoic Acid related orphan receptor alpha) gene was significantly lower among subjects (27.3%, N = 54) as compared to controls (42.9%, OR:0.5, CI: 0.26-0.96, p value 0.035). However, the significance of both disappeared after Bonferroni correction. Among clinical factors female gender was significantly associated with lithium non-response. CONCLUSION Although conducting pharmaco-genomic studies with large sample size is a challenge for low and middle-income countries, future studies can help improve the long-term outcome of youth with EOBD.
Collapse
|
6
|
Datta A. A Chemical Tool for Guiding Li Therapy. ACS CENTRAL SCIENCE 2021; 7:1783-1786. [PMID: 34841052 PMCID: PMC8614096 DOI: 10.1021/acscentsci.1c01289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Affiliation(s)
- Ankona Datta
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1 Homi Bhabha Road, Colaba, Mumbai 400005, India
| |
Collapse
|
7
|
Cope ZA, Kenton JA, Minassian A, Martin MV, Perry W, Bundgaard C, Arnt J, van Enkhuizen J, Geyer MA, Young JW. Chronic antipsychotic treatment exerts limited effects on the mania-like behavior of dopamine transporter knockdown mice. Behav Brain Res 2021; 405:113167. [PMID: 33577882 PMCID: PMC10729608 DOI: 10.1016/j.bbr.2021.113167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bipolar disorder is a life-threatening disorder linked to dopamine transporter (DAT) polymorphisms, with reduced DAT levels seen in positron emission tomography and postmortem brains. AIMS The purpose of this study was to examine the effects of approved antipsychotics on DAT dysfunction-mediated mania behavior in mice. METHODS DAT knockdown mice received either D2-family receptor antagonist risperidone or asenapine and mania-related behaviors were assessed in the clinically-relevant behavioral pattern monitor to assess spontaneous exploration. RESULTS Chronic risperidone did not reverse mania-like behavior in DAT knockdown mice. Chronic asenapine reduced mania behavior but this effect was more pronounced in wild-type littermates than in DAT knockdown mice. CONCLUSION Taken together, these findings suggest that while acute antipsychotic treatment may be beneficial in management of bipolar mania, more targeted therapeutics may be necessary for long-term treatment. Specific investigation into DAT-targeting drugs could improve future treatment of bipolar mania.
Collapse
Affiliation(s)
- Zackary A Cope
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Johnny A Kenton
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Center of Excellence for Stress and Mental Health and Research Service, VA San Diego Healthcare System, United States
| | - Maureen V Martin
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - William Perry
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Christoffer Bundgaard
- H. Lundbeck A/S. Neuroscience Research, Ottiliavej 9, DK-2500, Copenhagen, Valby, Denmark
| | - Jørn Arnt
- Sunred Pharma Consulting, Solrød Strand, Denmark
| | - Jordy van Enkhuizen
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0804, La Jolla, CA, 92093-0804, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States.
| |
Collapse
|
8
|
Pharmacogenomics of Lithium Response in Bipolar Disorder. Pharmaceuticals (Basel) 2021; 14:ph14040287. [PMID: 33804842 PMCID: PMC8063790 DOI: 10.3390/ph14040287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/22/2022] Open
Abstract
Despite being the most widely studied mood stabilizer, researchers have not confirmed a mechanism for lithium’s therapeutic efficacy in Bipolar Disorder (BD). Pharmacogenomic applications may be clinically useful in the future for identifying lithium-responsive patients and facilitating personalized treatment. Six genome-wide association studies (GWAS) reviewed here present evidence of genetic variations related to lithium responsivity and side effect expression. Variants were found on genes regulating the glutamate system, including GAD-like gene 1 (GADL1) and GRIA2 gene, a mutually-regulated target of lithium. In addition, single nucleotide polymorphisms (SNPs) discovered on SESTD1 may account for lithium’s exceptional ability to permeate cell membranes and mediate autoimmune and renal effects. Studies also corroborated the importance of epigenetics and stress regulation on lithium response, finding variants on long, non-coding RNA genes and associations between response and genetic loading for psychiatric comorbidities. Overall, the precision medicine model of stratifying patients based on phenotype seems to derive genotypic support of a separate clinical subtype of lithium-responsive BD. Results have yet to be expounded upon and should therefore be interpreted with caution.
Collapse
|
9
|
Gharipour M, Barekatain M, Sung J, Emami N, Sadeghian L, Dianatkhah M, Sarrafzadegan N, Jahanfar S. The Epigenetic Overlap between Obesity and Mood Disorders: A Systematic Review. Int J Mol Sci 2020; 21:ijms21186758. [PMID: 32942585 PMCID: PMC7555814 DOI: 10.3390/ijms21186758] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 01/19/2023] Open
Abstract
(1) Background: Obesity and mood disorders are considered as the most prevalent morbidities in many countries. We suppose that epigenetic mechanisms may induce higher rates of obesity in subjects who suffer from mood disorders. In this systematic review, we focused on the potential roles of DNA methylation on mood disorders and obesity development. (2) Methods: This systematic review was conducted in accordance with the PRISMA statement and registered in Prospero. A systematic search was conducted in MEDLINE, Scopus, Web of Science, Cochrane Central database, EMBASE, and CINHAL. We also conducted a Grey literature search, such as Google Scholar. (3) Results: After deduplication, we identified 198 potentially related citations. Finally, ten unique studies met our inclusion criteria. We have found three overlap genes that show significant DNA methylation changes, both in obesity and depression. Pathway analysis interaction for TAPBP, BDNF, and SORBS2 confirmed the relation of these genes in both obesity and mood disorders. (4) Conclusions: While mechanisms linking both obesity and mood disorders to epigenetic response are still unknown, we have already known chronic inflammation induces a novel epigenetic program. As the results of gene enrichment, pathways analysis showed that TAPBP, BDNF, and SORBS2 linked together by inflammatory pathways. Hypermethylation in these genes might play a crucial rule in the co-occurrence of obesity and mood disorders.
Collapse
Affiliation(s)
- Mojgan Gharipour
- Isfahan Cardiovascular Research Center, Genetics and Epigenetics Department, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Majid Barekatain
- Department of Psychiatry, School of Medicine and Behavioral Science Research Center, Isfahan University of Medical Science, Isfahan 8174673461, Iran;
| | - Johoon Sung
- Department Public Health Science, Genome & Health Big Data, Seoul National University, Seoul 05649, Korea;
| | - Naghmeh Emami
- Research Department, Interventional Cardiology Research Center, Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Ladan Sadeghian
- Research Department, Hypertension Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Minoo Dianatkhah
- Research Department, Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Shayesteh Jahanfar
- MPH Program, School of Public Health, Central Michigan University, Mount Pleasant, MI 48859, USA
- Correspondence: ; Tel.: +98-313-611-5116; Fax: +98-313-611-5303
| |
Collapse
|
10
|
Najafi H, Totonchi M, Sadeghizadeh M. Predicted Cellular and Molecular Actions of Lithium in the Treatment of Bipolar Disorder: An In Silico Study. CNS Drugs 2020; 34:521-533. [PMID: 32306228 DOI: 10.1007/s40263-020-00723-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Lithium remains the first-line treatment for bipolar disorder (BD), but patients respond to it variably. While a myriad of studies have attributed many genes and signaling pathways to lithium responsiveness, a comprehensive study with an integrated conclusion is still lacking. OBJECTIVE We aim to present an integrated mechanism for the therapeutic actions of lithium in BD. METHODS First, a list of lithium responsiveness-associated genes (LRAGs) was collected by searching in the literature. Thereafter, gene set enrichment analysis together with gene-gene interaction network analysis was performed, in order to find the cellular and molecular events related to the LRAGs. RESULTS Gene set enrichment analyses showed that the chromosomal regions 3p26, 4p21, 5q34 and 7p13 could be novel associated loci for lithium responsiveness in BD. Also, expression pattern analysis of the LRAGs showed their enrichment in adulthood stages and different cell lineages of brain, blood and immune system. Most of the LRAGs exhibited enriched expression in central parts of human brain, suggesting major contribution of these parts in lithium responsiveness. Beside the prediction of several biological processes and signaling pathways related to lithium responsiveness, an interaction network between these processes was constructed that was found to be regulated by a set of microRNAs. Proteins of the network were mainly classified as transcription factors and kinases, which also highlighted the crucial role of glycogen synthase kinase 3β (GSK3β) in lithium responsiveness. CONCLUSIONS The predicted cellular and molecular events in this study could be considered as mechanisms and also determinants of lithium responsiveness in BD.
Collapse
Affiliation(s)
- Hadi Najafi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran.
| |
Collapse
|
11
|
Stern S, Linker S, Vadodaria KC, Marchetto MC, Gage FH. Prediction of Response to Drug Therapy in Psychiatric Disorders. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2019; 17:294-307. [PMID: 32015721 PMCID: PMC6996058 DOI: 10.1176/appi.focus.17304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Reprinted with permission from Open Biol. 8: 180031. The Royal Society.
Collapse
|
12
|
Stern S, Linker S, Vadodaria KC, Marchetto MC, Gage FH. Prediction of response to drug therapy in psychiatric disorders. Open Biol 2019; 8:rsob.180031. [PMID: 29794033 PMCID: PMC5990649 DOI: 10.1098/rsob.180031] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/02/2018] [Indexed: 12/20/2022] Open
Abstract
Personalized medicine has become increasingly relevant to many medical fields, promising more efficient drug therapies and earlier intervention. The development of personalized medicine is coupled with the identification of biomarkers and classification algorithms that help predict the responses of different patients to different drugs. In the last 10 years, the Food and Drug Administration (FDA) has approved several genetically pre-screened drugs labelled as pharmacogenomics in the fields of oncology, pulmonary medicine, gastroenterology, haematology, neurology, rheumatology and even psychiatry. Clinicians have long cautioned that what may appear to be similar patient-reported symptoms may actually arise from different biological causes. With growing populations being diagnosed with different psychiatric conditions, it is critical for scientists and clinicians to develop precision medication tailored to individual conditions. Genome-wide association studies have highlighted the complicated nature of psychiatric disorders such as schizophrenia, bipolar disorder, major depression and autism spectrum disorder. Following these studies, association studies are needed to look for genomic markers of responsiveness to available drugs of individual patients within the population of a specific disorder. In addition to GWAS, the advent of new technologies such as brain imaging, cell reprogramming, sequencing and gene editing has given us the opportunity to look for more biomarkers that characterize a therapeutic response to a drug and to use all these biomarkers for determining treatment options. In this review, we discuss studies that were performed to find biomarkers of responsiveness to different available drugs for four brain disorders: bipolar disorder, schizophrenia, major depression and autism spectrum disorder. We provide recommendations for using an integrated method that will use available techniques for a better prediction of the most suitable drug.
Collapse
Affiliation(s)
- Shani Stern
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sara Linker
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Krishna C Vadodaria
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Maria C Marchetto
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
The identification of biomarkers predicting acute and maintenance lithium treatment response in bipolar disorder: A plea for further research attention. Psychiatry Res 2018; 269:658-672. [PMID: 30216918 DOI: 10.1016/j.psychres.2018.08.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/19/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
The prediction of acute and maintenance lithium treatment response carries major clinical and neurobiological implications, warranting systematic review. A Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) compliant review searched major electronic databases from inception until December 2017 for studies documenting a clinical diagnosis of bipolar disorder (BD) made according to the mainstream diagnostic manuals and confirmed by a structured interview. Eligible studies allowed a quantitative comparison of endpoint vs baseline mean values of a given biomarker, regardless of the mood phase of patients with BD, and the disorder was assessed for severity using validated rating tool(s). Owing to the purposely applied stringent selection criteria, 16 acute and 12 maintenance studies could be included. The anticipated publication bias limited the chances of reportable generalizable findings, hindering a side-by-side comparison of different records across varying biomarkers and subsequent meta-analyses. The PRISMA approach was nonetheless preferred; it aimed at enhancing the homogeneity of the included results and minimizing the chances of "apples and oranges" with respect to the present research theme. The present critical review confirms the need for future research to specifically assess either pretreatment and/or posttreatment putative biomarkers of patients with BD and treated with lithium.
Collapse
|
14
|
Genetic or pharmacological superoxide-hydrogen peroxide imbalances modulate the in vitro effects of lithium on glycogen synthase kinase-3β. Gene 2018; 655:48-55. [DOI: 10.1016/j.gene.2018.02.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 11/20/2022]
|
15
|
Fortinguerra S, Buriani A, Sorrenti V, Lenzi M, Giusti P. Molecular network-selected pharmacogenomics in a case of bipolar spectrum disorder. Pharmacogenomics 2017; 18:1631-1642. [PMID: 29173093 DOI: 10.2217/pgs-2017-0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Personal genomic analysis was used for molecular diagnosis and pharmacogenomics in a 53-year-old female suffering from alternating depressive and dysphoric episodes. A total of 52 genes and 108 SNPs were analyzed in the whole genome. Results from the pharmacogenomic analysis were consistent with the pharmacological history and indicate mutations associated with low monoaminergic tone, but also a hyperactive 5HT2A receptor, a feature that associates to a high probability of developing a bipolar condition, especially under 5-hydroxytryptamine potentiating pharmacology. This aligns with the patient developing dysphoria with high clomipramine. The pharmacokinetic genomics pointed out to some absorption, distribution, metabolism, and excretion (ADME) alterations that can lower or nullify drug's activity. A personalized regimen was proposed, with a positive outcome after 1 year.
Collapse
Affiliation(s)
- Stefano Fortinguerra
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, (Synlab Limited) Padova 35100, Italy
| | - Alessandro Buriani
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, (Synlab Limited) Padova 35100, Italy
| | - Vincenzo Sorrenti
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, (Synlab Limited) Padova 35100, Italy.,Department of Pharmaceutical & Pharmacological Sciences, University of Padova, Padova, Italy
| | - Michele Lenzi
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group, (Synlab Limited) Padova 35100, Italy
| | - Pietro Giusti
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
16
|
Budde M, Degner D, Brockmöller J, Schulze TG. Pharmacogenomic aspects of bipolar disorder: An update. Eur Neuropsychopharmacol 2017; 27:599-609. [PMID: 28342679 DOI: 10.1016/j.euroneuro.2017.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 01/31/2017] [Accepted: 02/09/2017] [Indexed: 12/11/2022]
Abstract
The hopes for readily implementable precision medicine are high. For many complex disorders, such as bipolar disorder, these hopes critically hinge on tangible successes in pharmacogenetics of treatment response or susceptibility to adverse events. In this article, we review the current state of pharmacogenomics of bipolar disorder including latest results from candidate genes and genome-wide association studies. The majority of studies focus on response to lithium treatment. Although a host of genes has been studied, hardly any replicated findings have emerged so far. Very small samples sizes and heterogeneous phenotype definition may be considered the major impediments to success in this field. Drawing from current experiences and successes in studies on diagnostic psychiatric phenotypes, we suggest several approaches for our way forward.
Collapse
Affiliation(s)
- M Budde
- Institute of Psychiatric Phenomics and Genomics, Clinical Center of the University of Munich, Nussbaumstr. 7, 80336 Munich, Germany; University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - D Degner
- University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - J Brockmöller
- University Medical Center Göttingen, Department of Clinical Pharmacology, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - T G Schulze
- Institute of Psychiatric Phenomics and Genomics, Clinical Center of the University of Munich, Nussbaumstr. 7, 80336 Munich, Germany; University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| |
Collapse
|
17
|
Genome-wide association study identifies SESTD1 as a novel risk gene for lithium-responsive bipolar disorder. Mol Psychiatry 2016; 21:1290-7. [PMID: 26503763 PMCID: PMC4995544 DOI: 10.1038/mp.2015.165] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/04/2015] [Accepted: 09/24/2015] [Indexed: 12/02/2022]
Abstract
Lithium is the mainstay prophylactic treatment for bipolar disorder (BD), but treatment response varies considerably across individuals. Patients who respond well to lithium treatment might represent a relatively homogeneous subtype of this genetically and phenotypically diverse disorder. Here, we performed genome-wide association studies (GWAS) to identify (i) specific genetic variations influencing lithium response and (ii) genetic variants associated with risk for lithium-responsive BD. Patients with BD and controls were recruited from Sweden and the United Kingdom. GWAS were performed on 2698 patients with subjectively defined (self-reported) lithium response and 1176 patients with objectively defined (clinically documented) lithium response. We next conducted GWAS comparing lithium responders with healthy controls (1639 subjective responders and 8899 controls; 323 objective responders and 6684 controls). Meta-analyses of Swedish and UK results revealed no significant associations with lithium response within the bipolar subjects. However, when comparing lithium-responsive patients with controls, two imputed markers attained genome-wide significant associations, among which one was validated in confirmatory genotyping (rs116323614, P=2.74 × 10(-8)). It is an intronic single-nucleotide polymorphism (SNP) on chromosome 2q31.2 in the gene SEC14 and spectrin domains 1 (SESTD1), which encodes a protein involved in regulation of phospholipids. Phospholipids have been strongly implicated as lithium treatment targets. Furthermore, we estimated the proportion of variance for lithium-responsive BD explained by common variants ('SNP heritability') as 0.25 and 0.29 using two definitions of lithium response. Our results revealed a genetic variant in SESTD1 associated with risk for lithium-responsive BD, suggesting that the understanding of BD etiology could be furthered by focusing on this subtype of BD.
Collapse
|
18
|
Bush WS, Crosslin DR, Owusu‐Obeng A, Wallace J, Almoguera B, Basford MA, Bielinski SJ, Carrell DS, Connolly JJ, Crawford D, Doheny KF, Gallego CJ, Gordon AS, Keating B, Kirby J, Kitchner T, Manzi S, Mejia AR, Pan V, Perry CL, Peterson JF, Prows CA, Ralston J, Scott SA, Scrol A, Smith M, Stallings SC, Veldhuizen T, Wolf W, Volpi S, Wiley K, Li R, Manolio T, Bottinger E, Brilliant MH, Carey D, Chisholm RL, Chute CG, Haines JL, Hakonarson H, Harley JB, Holm IA, Kullo IJ, Jarvik GP, Larson EB, McCarty CA, Williams MS, Denny JC, Rasmussen‐Torvik LJ, Roden DM, Ritchie MD. Genetic variation among 82 pharmacogenes: The PGRNseq data from the eMERGE network. Clin Pharmacol Ther 2016; 100:160-9. [PMID: 26857349 PMCID: PMC5010878 DOI: 10.1002/cpt.350] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/12/2016] [Accepted: 02/04/2016] [Indexed: 12/20/2022]
Abstract
Genetic variation can affect drug response in multiple ways, although it remains unclear how rare genetic variants affect drug response. The electronic Medical Records and Genomics (eMERGE) Network, collaborating with the Pharmacogenomics Research Network, began eMERGE‐PGx, a targeted sequencing study to assess genetic variation in 82 pharmacogenes critical for implementation of “precision medicine.” The February 2015 eMERGE‐PGx data release includes sequence‐derived data from ∼5,000 clinical subjects. We present the variant frequency spectrum categorized by variant type, ancestry, and predicted function. We found 95.12% of genes have variants with a scaled Combined Annotation‐Dependent Depletion score above 20, and 96.19% of all samples had one or more Clinical Pharmacogenetics Implementation Consortium Level A actionable variants. These data highlight the distribution and scope of genetic variation in relevant pharmacogenes, identifying challenges associated with implementing clinical sequencing for drug treatment at a broader level, underscoring the importance for multifaceted research in the execution of precision medicine.
Collapse
|
19
|
Landgraf D, Joiner WJ, McCarthy MJ, Kiessling S, Barandas R, Young JW, Cermakian N, Welsh DK. The mood stabilizer valproic acid opposes the effects of dopamine on circadian rhythms. Neuropharmacology 2016; 107:262-270. [PMID: 27033596 DOI: 10.1016/j.neuropharm.2016.03.047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/17/2016] [Accepted: 03/25/2016] [Indexed: 12/21/2022]
Abstract
Endogenous circadian (∼24 h) clocks regulate key physiological and cognitive processes via rhythmic expression of clock genes. The main circadian pacemaker is the hypothalamic suprachiasmatic nucleus (SCN). Mood disorders, including bipolar disorder (BD), are commonly associated with disturbed circadian rhythms. Dopamine (DA) contributes to mania in BD and has direct impact on clock gene expression. Therefore, we hypothesized that high levels of DA during episodes of mania contribute to disturbed circadian rhythms in BD. The mood stabilizer valproic acid (VPA) also affects circadian rhythms. Thus, we further hypothesized that VPA normalizes circadian disturbances caused by elevated levels of DA. To test these hypotheses, we examined locomotor rhythms and circadian gene cycling in mice with reduced expression of the dopamine transporter (DAT-KD mice), which results in elevated DA levels and mania-like behavior. We found that elevated DA signaling lengthened the circadian period of behavioral rhythms in DAT-KD mice and clock gene expression rhythms in SCN explants. In contrast, we found that VPA shortened circadian period of behavioral rhythms in DAT-KD mice and clock gene expression rhythms in SCN explants, hippocampal cell lines, and human fibroblasts from BD patients. Thus, DA and VPA have opposing effects on circadian period. To test whether the impact of VPA on circadian rhythms contributes to its behavioral effects, we fed VPA to DAT-deficient Drosophila with and without functioning circadian clocks. Consistent with our hypothesis, we found that VPA had potent activity-suppressing effects in hyperactive DAT-deficient flies with intact circadian clocks. However, these effects were attenuated in DAT-deficient flies in which circadian clocks were disrupted, suggesting that VPA functions partly through the circadian clock to suppress activity. Here, we provide in vivo and in vitro evidence across species that elevated DA signaling lengthens the circadian period, an effect remediated by VPA treatment. Hence, VPA may exert beneficial effects on mood by normalizing lengthened circadian rhythm period in subjects with elevated DA resulting from reduced DAT.
Collapse
Affiliation(s)
- Dominic Landgraf
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA.
| | - William J Joiner
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Michael J McCarthy
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Silke Kiessling
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Rita Barandas
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Psychiatry, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal; Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Jared W Young
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Nicolas Cermakian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - David K Welsh
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
Hou L, Heilbronner U, Degenhardt F, Adli M, Akiyama K, Akula N, Ardau R, Arias B, Backlund L, Banzato CEM, Benabarre A, Bengesser S, Bhattacharjee AK, Biernacka JM, Birner A, Brichant-Petitjean C, Bui ET, Cervantes P, Chen GB, Chen HC, Chillotti C, Cichon S, Clark SR, Colom F, Cousins DA, Cruceanu C, Czerski PM, Dantas CR, Dayer A, Étain B, Falkai P, Forstner AJ, Frisén L, Fullerton JM, Gard S, Garnham JS, Goes FS, Grof P, Gruber O, Hashimoto R, Hauser J, Herms S, Hoffmann P, Hofmann A, Jamain S, Jiménez E, Kahn JP, Kassem L, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Lackner N, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, Jaramillo CAL, MacQueen G, Manchia M, Martinsson L, Mattheisen M, McCarthy MJ, McElroy SL, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Nöthen MM, Ösby U, Ozaki N, Perlis RH, Pfennig A, Reich-Erkelenz D, Rouleau GA, Schofield PR, Schubert KO, Schweizer BW, Seemüller F, Severino G, Shekhtman T, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Smoller JW, Squassina A, Stamm T, Stopkova P, Tighe SK, Tortorella A, Turecki G, Volkert J, Witt S, Wright A, Young LT, Zandi PP, Potash JB, DePaulo JR, et alHou L, Heilbronner U, Degenhardt F, Adli M, Akiyama K, Akula N, Ardau R, Arias B, Backlund L, Banzato CEM, Benabarre A, Bengesser S, Bhattacharjee AK, Biernacka JM, Birner A, Brichant-Petitjean C, Bui ET, Cervantes P, Chen GB, Chen HC, Chillotti C, Cichon S, Clark SR, Colom F, Cousins DA, Cruceanu C, Czerski PM, Dantas CR, Dayer A, Étain B, Falkai P, Forstner AJ, Frisén L, Fullerton JM, Gard S, Garnham JS, Goes FS, Grof P, Gruber O, Hashimoto R, Hauser J, Herms S, Hoffmann P, Hofmann A, Jamain S, Jiménez E, Kahn JP, Kassem L, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Lackner N, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, Jaramillo CAL, MacQueen G, Manchia M, Martinsson L, Mattheisen M, McCarthy MJ, McElroy SL, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Nöthen MM, Ösby U, Ozaki N, Perlis RH, Pfennig A, Reich-Erkelenz D, Rouleau GA, Schofield PR, Schubert KO, Schweizer BW, Seemüller F, Severino G, Shekhtman T, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Smoller JW, Squassina A, Stamm T, Stopkova P, Tighe SK, Tortorella A, Turecki G, Volkert J, Witt S, Wright A, Young LT, Zandi PP, Potash JB, DePaulo JR, Bauer M, Reininghaus EZ, Novák T, Aubry JM, Maj M, Baune BT, Mitchell PB, Vieta E, Frye MA, Rybakowski JK, Kuo PH, Kato T, Grigoroiu-Serbanescu M, Reif A, Del Zompo M, Bellivier F, Schalling M, Wray NR, Kelsoe JR, Alda M, Rietschel M, McMahon FJ, Schulze TG. Genetic variants associated with response to lithium treatment in bipolar disorder: a genome-wide association study. Lancet 2016; 387:1085-1093. [PMID: 26806518 PMCID: PMC4814312 DOI: 10.1016/s0140-6736(16)00143-4] [Show More Authors] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lithium is a first-line treatment in bipolar disorder, but individual response is variable. Previous studies have suggested that lithium response is a heritable trait. However, no genetic markers of treatment response have been reproducibly identified. METHODS Here, we report the results of a genome-wide association study of lithium response in 2563 patients collected by 22 participating sites from the International Consortium on Lithium Genetics (ConLiGen). Data from common single nucleotide polymorphisms (SNPs) were tested for association with categorical and continuous ratings of lithium response. Lithium response was measured using a well established scale (Alda scale). Genotyped SNPs were used to generate data at more than 6 million sites, using standard genomic imputation methods. Traits were regressed against genotype dosage. Results were combined across two batches by meta-analysis. FINDINGS A single locus of four linked SNPs on chromosome 21 met genome-wide significance criteria for association with lithium response (rs79663003, p=1·37 × 10(-8); rs78015114, p=1·31 × 10(-8); rs74795342, p=3·31 × 10(-9); and rs75222709, p=3·50 × 10(-9)). In an independent, prospective study of 73 patients treated with lithium monotherapy for a period of up to 2 years, carriers of the response-associated alleles had a significantly lower rate of relapse than carriers of the alternate alleles (p=0·03268, hazard ratio 3·8, 95% CI 1·1-13·0). INTERPRETATION The response-associated region contains two genes for long, non-coding RNAs (lncRNAs), AL157359.3 and AL157359.4. LncRNAs are increasingly appreciated as important regulators of gene expression, particularly in the CNS. Confirmed biomarkers of lithium response would constitute an important step forward in the clinical management of bipolar disorder. Further studies are needed to establish the biological context and potential clinical utility of these findings. FUNDING Deutsche Forschungsgemeinschaft, National Institute of Mental Health Intramural Research Program.
Collapse
Affiliation(s)
- Liping Hou
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich, Munich, Germany; Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August University Göttingen, Göttingen, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany; Fliedner Klinik Berlin, Center for Psychiatry, Psychotherapy and Psychosomatic Medicine, Berlin, Germany
| | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Nirmala Akula
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, Hospital University Agency of Cagliari, Cagliari, Italy
| | - Bárbara Arias
- Department of Biologia Animal, Unitat d'Antropologia (Dp Biología Animal), Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, CIBERSAM, Barcelona, Catalonia, Spain
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Claudio E M Banzato
- Department of Psychiatry, University of Campinas (Unicamp), Campinas, Brazil
| | - Antoni Benabarre
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Susanne Bengesser
- Neurobiological Background and Anthropometrics in Bipolar Affective Disorder, Department of Psychiatry, Medical University of Graz, Graz, Austria
| | | | - Joanna M Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Armin Birner
- Neurobiological Background and Anthropometrics in Bipolar Affective Disorder, Department of Psychiatry, Medical University of Graz, Graz, Austria
| | - Clara Brichant-Petitjean
- INSERM UMR-S 1144-Université Paris Diderot Pôle de Psychiatrie, AP-HP, Groupe Hospitalier Lariboisière-F Widal, Paris, France
| | - Elise T Bui
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Pablo Cervantes
- The Neuromodulation Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Guo-Bo Chen
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland, QLD, Australia
| | - Hsi-Chung Chen
- Department of Psychiatry & Center of Sleep Disorders, National Taiwan University Hospital, Taipei, Taiwan
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, Hospital University Agency of Cagliari, Cagliari, Italy
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany; Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Scott R Clark
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Francesc Colom
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - David A Cousins
- Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Cristiana Cruceanu
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Piotr M Czerski
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Clarissa R Dantas
- Department of Psychiatry, University of Campinas (Unicamp), Campinas, Brazil
| | - Alexandre Dayer
- Department of Mental Health and Psychiatry, Mood Disorders Unit-Geneva University Hospitals, Geneva, Switzerland
| | - Bruno Étain
- Inserm U955, Psychiatrie Translationnelle, Créteil, France
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Louise Frisén
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Child and Adolescent Psychiatry Research Center, Stockholm, Sweden
| | - Janice M Fullerton
- Mental Illness Research Theme, Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Sébastien Gard
- Pôle de Psychiatrie Générale Universitaire, Centre Hospitalier Charles Perrens, Bordeaux, France
| | - Julie S Garnham
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, NS, Canada
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Paul Grof
- Mood Disorders Center of Ottawa, Ottawa, ON, Canada
| | - Oliver Gruber
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August University Göttingen, Göttingen, Germany
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany; Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany; Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Andrea Hofmann
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | | | - Esther Jiménez
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Jean-Pierre Kahn
- Service de Psychiatrie et Psychologie Clinique, Centre Psychothérapique de Nancy-Laxou-Université de Lorraine, Nancy, France
| | - Layla Kassem
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Sebastian Kliwicki
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara König
- Department of Psychiatry and Psychotherapeutic Medicine, Landesklinikum Neunkirchen, Neunkirchen, Austria
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nina Lackner
- Neurobiological Background and Anthropometrics in Bipolar Affective Disorder, Department of Psychiatry, Medical University of Graz, Graz, Austria
| | - Gonzalo Laje
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Mikael Landén
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the Gothenburg University, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Marion Leboyer
- Assistance Publique-Hôpitaux de Paris, Hôpital Albert Chenevier-Henri Mondor, Pôle de Psychiatrie, Créteil, France
| | - Susan G Leckband
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Pharmacy, VA San Diego Healthcare System, San Diego, CA, USA
| | | | - Glenda MacQueen
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Mirko Manchia
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Lina Martinsson
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael J McCarthy
- Department of Psychiatry, VA San Diego Healthcare System, San Diego, CA, USA
| | - Susan L McElroy
- Department of Psychiatry, Lindner Center of Hope, University of Cincinnati, Mason, OH, USA
| | - Marina Mitjans
- Department of Biologia Animal, Unitat d'Antropologia (Dp Biología Animal), Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, CIBERSAM, Barcelona, Catalonia, Spain
| | - Francis M Mondimore
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Palmiero Monteleone
- Neurosciences Section, Department of Medicine and Surgery, University of Salerno, Salerno, Italy; Department of Psychiatry, University of Naples SUN, Naples, Italy
| | | | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Urban Ösby
- Department of Neurobiology, Care sciences, and Society, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Roy H Perlis
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrea Pfennig
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Daniela Reich-Erkelenz
- Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Guy A Rouleau
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Peter R Schofield
- Mental Illness Research Theme, Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Barbara W Schweizer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Florian Seemüller
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Giovanni Severino
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Tatyana Shekhtman
- Veterans Administration, San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Paul D Shilling
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Kazutaka Shimoda
- Department of Psychiatry, Dokkyo Medical University School of Medicine, Mibu, Japan
| | | | - Claire M Slaney
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, NS, Canada
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alessio Squassina
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Thomas Stamm
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Pavla Stopkova
- National Institute of Mental Health, Klecany, Czech Republic
| | - Sarah K Tighe
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA; University of Iowa Carver College of Medicine and University of Iowa College of Public Health, VA Quality Scholars Program, Iowa City VA Hospital, Iowa City, IA, USA
| | | | - Gustavo Turecki
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Julia Volkert
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Stephanie Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adam Wright
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - L Trevor Young
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James B Potash
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - J Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Eva Z Reininghaus
- Neurobiological Background and Anthropometrics in Bipolar Affective Disorder, Department of Psychiatry, Medical University of Graz, Graz, Austria
| | - Tomas Novák
- National Institute of Mental Health, Klecany, Czech Republic
| | - Jean-Michel Aubry
- Department of Mental Health and Psychiatry, Mood Disorders Unit-Geneva University Hospitals, Geneva, Switzerland
| | - Mario Maj
- Department of Psychiatry, University of Naples SUN, Naples, Italy
| | - Bernhard T Baune
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Philip B Mitchell
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - Eduard Vieta
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Janusz K Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Saitama, Japan
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Psychiatric Hospital, Bucharest, Romania
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Maria Del Zompo
- Unit of Clinical Pharmacology, Hospital University Agency of Cagliari, Cagliari, Italy; Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Frank Bellivier
- INSERM UMR-S 1144-Université Paris Diderot Pôle de Psychiatrie, AP-HP, Groupe Hospitalier Lariboisière-F Widal, Paris, France
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Naomi R Wray
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland, QLD, Australia
| | - John R Kelsoe
- Veterans Administration, San Diego Healthcare System, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, NS, Canada; National Institute of Mental Health, Klecany, Czech Republic
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Francis J McMahon
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA.
| | - Thomas G Schulze
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA; Institute of Psychiatric Phenomics and Genomics, Ludwig-Maximilians-University Munich, Munich, Germany; Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August University Göttingen, Göttingen, Germany; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
21
|
Exploring Genetic Variability at PI, GSK3, HPA, and Glutamatergic Pathways in Lithium Response: Association With IMPA2, INPP1, and GSK3B Genes. J Clin Psychopharmacol 2015; 35:600-4. [PMID: 26267417 DOI: 10.1097/jcp.0000000000000382] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lithium is considered the first-line treatment in bipolar disorder, although response could range from an excellent response to a complete lack of response. Response to lithium is a complex phenotype in which different factors, part of them genetics, are involved. In this sense, the aim of this study was to investigate the potential association of genetic variability at genes related to phosphoinositide, glycogen synthetase kinase-3 (GSK3), hypothalamic-pituitary-adrenal, and glutamatergic pathways with lithium response. A sample of 131 bipolar patients (99 type I, 32 type II) were grouped and compared according to their level of response: excellent responders (ER), partial responders (PR), and nonresponders (NR). Genotype and allele distributions of the rs669838 (IMPA2), rs909270 (INNP1), rs11921360 (GSK3B), and rs28522620 (GRIK2) polymorphisms significantly differed between ER, PR, and NR. When we compared the ER versus PR+NR, the logistic regression showed significant association for rs669838-C (IMPA2; P = 0.021), rs909270-G (INPP1; P = 0.009), and rs11921360-A (GSK3B; P = 0.004) with lithium nonresponse. Haplotype analysis showed significant association for the haplotypes rs3791809-rs4853694-rs909270 (INPP1) and rs1732170-rs11921360-rs334558 (GSK3B) and lithium response. Our study is in line with previous studies reporting association between genetic variability at these genes and lithium response, pointing to an effect of IMPA2, INPP1, and GSK3B genes to lithium response in bipolar disorder patients. Further studies with larger samples are warranted to assess the strength of the reported associations.
Collapse
|
22
|
Cellular models to study bipolar disorder: A systematic review. J Affect Disord 2015; 184:36-50. [PMID: 26070045 DOI: 10.1016/j.jad.2015.05.037] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is an emerging interest in the use of cellular models to study psychiatric disorders. We have systematically reviewed the application of cellular models to understand the biological basis of bipolar disorder (BD). METHOD Published scientific literature in MEDLINE, PsychINFO and SCOPUS databases were identified with the following search strategy: [(Lymphoblastoid OR Lymphoblast OR Fibroblast OR Pluripotent OR Olfactory epithelium OR Olfactory mucosa) AND (Bipolar disorder OR Lithium OR Valproate OR Mania)]. Studies were included if they had used cell cultures derived from BD patients. RESULTS There were 65 articles on lymphoblastoid cell lines, 14 articles on fibroblasts, 4 articles on olfactory neuronal epithelium (ONE) and 2 articles on neurons reprogrammed from induced pluripotent stem cell lines (IPSC). Several parameters have been studied, and the most replicated findings are abnormalities in calcium signaling, endoplasmic reticulum (ER) stress response, mitochondrial oxidative pathway, membrane ion channels, circadian system and apoptosis related genes. These, although present in basal state, seem to be accentuated in the presence of cellular stressors (e.g. oxidative stress--rotenone; ER stress--thapsigargin), and are often reversed with in-vitro lithium. CONCLUSION Cellular modeling has proven useful in BD, and potential pathways, especially in cellular resilience related mechanisms have been identified. These findings show consistency with other study designs (genome-wide association, brain-imaging, and post-mortem brain expression). ONE cells and IPSC reprogrammed neurons represent the next generation of cell models in BD. Future studies should focus on family-based study designs and combine cell models with deep sequencing and genetic manipulations.
Collapse
|
23
|
Higgins GA, Allyn-Feuer A, Barbour E, Athey BD. A glutamatergic network mediates lithium response in bipolar disorder as defined by epigenome pathway analysis. Pharmacogenomics 2015; 16:1547-63. [PMID: 26343379 DOI: 10.2217/pgs.15.106] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM A regulatory network in the human brain mediating lithium response in bipolar patients was revealed by analysis of functional SNPs from genome-wide association studies (GWAS) and published gene association studies, followed by epigenome mapping. METHODS An initial set of 23,312 SNPs in linkage disequilibrium with lead SNPs, and sub-threshold GWAS SNPs rescued by pathway analysis, were studied in the same populations. These were assessed using our workflow and annotation by the epigenome roadmap consortium. RESULTS Twenty-seven percent of 802 SNPs that were associated with lithium response (13 published studies gene association studies and two GWAS) were shared in common with 1281 SNPs from 18 GWAS examining psychiatric disorders and adverse events associated with lithium treatment. Nineteen SNPs were annotated as active regulatory elements such as enhancers and promoters in a tissue-specific manner. They were located within noncoding regions of ten genes: ANK3, ARNTL, CACNA1C, CACNG2, CDKN1A, CREB1, GRIA2, GSK3B, NR1D1 and SLC1A2. Following gene set enrichment and pathway analysis, these genes were found to be significantly associated (p = 10(-27); Fisher exact test) with an AMPA2 glutamate receptor network in human brain. Our workflow results showed concordance with annotation of regulatory elements from the epigenome roadmap. Analysis of cognate mRNA and enhancer RNA exhibited patterns consistent with an integrated pathway in human brain. CONCLUSION This pharmacoepigenomic regulatory pathway is located in the same brain regions that exhibit tissue volume loss in bipolar disorder. Although in silico analysis requires biological validation, the approach provides value for identification of candidate variants that may be used in pharmacogenomic testing to identify bipolar patients likely to respond to lithium.
Collapse
Affiliation(s)
- Gerald A Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Pharmacogenomic Science, Assurex Health, Inc., Mason, OH 45040, USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Edward Barbour
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brian D Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
24
|
Lithium in the treatment of bipolar disorder: pharmacology and pharmacogenetics. Mol Psychiatry 2015; 20:661-70. [PMID: 25687772 PMCID: PMC5125816 DOI: 10.1038/mp.2015.4] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/22/2014] [Accepted: 12/19/2014] [Indexed: 01/09/2023]
Abstract
After decades of research, the mechanism of action of lithium in preventing recurrences of bipolar disorder remains only partially understood. Lithium research is complicated by the absence of suitable animal models of bipolar disorder and by having to rely on in vitro studies of peripheral tissues. A number of distinct hypotheses emerged over the years, but none has been conclusively supported or rejected. The common theme emerging from pharmacological and genetic studies is that lithium affects multiple steps in cellular signaling, usually enhancing basal and inhibiting stimulated activities. Some of the key nodes of these regulatory networks include GSK3 (glycogen synthase kinase 3), CREB (cAMP response element-binding protein) and Na(+)-K(+) ATPase. Genetic and pharmacogenetic studies are starting to generate promising findings, but remain limited by small sample sizes. As full responders to lithium seem to represent a unique clinical population, there is inherent value and need for studies of lithium responders. Such studies will be an opportunity to uncover specific effects of lithium in those individuals who clearly benefit from the treatment.
Collapse
|
25
|
Lewitzka U, Jabs B, Fülle M, Holthoff V, Juckel G, Uhl I, Kittel-Schneider S, Reif A, Reif-Leonhard C, Gruber O, Djawid B, Goodday S, Haussmann R, Pfennig A, Ritter P, Conell J, Severus E, Bauer M. Does lithium reduce acute suicidal ideation and behavior? A protocol for a randomized, placebo-controlled multicenter trial of lithium plus Treatment As Usual (TAU) in patients with suicidal major depressive episode. BMC Psychiatry 2015; 15:117. [PMID: 25986590 PMCID: PMC4458032 DOI: 10.1186/s12888-015-0499-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/15/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lithium has proven suicide preventing effects in the long-term treatment of patients with affective disorders. Clinical evidence from case reports indicate that this effect may occur early on at the beginning of lithium treatment. The impact of lithium treatment on acute suicidal thoughts and/or behavior has not been systematically studied in a controlled trial. The primary objective of this confirmatory study is to determine the association between lithium therapy and acute suicidal ideation and/or suicidal behavior in inpatients with a major depressive episode (MDE, unipolar and bipolar disorder according to DSM IV criteria). The specific aim is to test the hypothesis that lithium plus treatment as usual (TAU), compared to placebo plus TAU, results in a significantly greater decrease in suicidal ideation and/or behavior over 5 weeks in inpatients with MDE. METHODS/DESIGN We initiated a randomized, placebo-controlled multicenter trial. Patients with the diagnosis of a moderate to severe depressive episode and suicidal thoughts and/or suicidal behavior measured with the Sheehan-Suicidality-Tracking Scale (S-STS) will be randomly allocated to add lithium or placebo to their treatment as usual. Change in the clinician administered S-STS from the initial to the final visit will be the primary outcome. DISCUSSION There is an urgent need to identify treatments that will acutely decrease suicidal ideation and/or suicidal behavior. The results of this study will demonstrate whether lithium reduces suicidal ideation and behavior within the first 5 weeks of treatment. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02039479.
Collapse
Affiliation(s)
- U. Lewitzka
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - B. Jabs
- Psychiatric Department of the Municipal Hospital Dresden-Neustadt, Dresden, Germany
| | - M. Fülle
- Psychiatric Department of the Municipal Hospital Dresden-Neustadt, Dresden, Germany
| | - V. Holthoff
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine, Alexianer Krankenhaus Hedwigshöhe, Berlin, Germany
| | - G. Juckel
- Department of Psychiatry, Psychotherapy, Prevention Medicine, LWL-University Clinic Bochum, Ruhr University Bochum, Bochum, Germany
| | - I. Uhl
- Department of Psychiatry, Psychotherapy, Prevention Medicine, LWL-University Clinic Bochum, Ruhr University Bochum, Bochum, Germany
| | - S. Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe-Universit, Frankfurt, Germany
| | - A. Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe-Universit, Frankfurt, Germany
| | - C. Reif-Leonhard
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe-Universit, Frankfurt, Germany
| | - O. Gruber
- Center for Translational Research in Systems Neuroscience and Psychiatry, Clinic for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - B. Djawid
- Coordination Centre for Clinical, Trials (KKS) Dresden, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden
, Germany
| | - S. Goodday
- Department of Epidemiology Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - R. Haussmann
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - A. Pfennig
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - P. Ritter
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - J. Conell
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - E. Severus
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| | - M. Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, Dresden, D-01307 Germany
| |
Collapse
|
26
|
Drago A, Monti B, De Ronchi D, Serretti A. CRY1 Variations Impacts on the Depressive Relapse Rate in a Sample of Bipolar Patients. Psychiatry Investig 2015; 12:118-24. [PMID: 25670954 PMCID: PMC4310909 DOI: 10.4306/pi.2015.12.1.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/31/2013] [Accepted: 12/31/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE A relevant part of the social and personal burden caused by Bipolar Disorder (BD) is related to depressive phases. Authors investigated the genetic impact of a set of variations located in CRY1, a gene involved in the control of the circadian rhythms, towards depressive episodes in a sample of bipolar patients from the STEP-BD sample. As a secondary analysis, CYR1 variations were analyzed as predictors of sleep disruption. METHODS 654 bipolar patients were included in the analysis. Data were available genome-wide. The part of the genome coding for the CRY1 was imputed and pruned according to standards in the field. 7 SNPs were available for the analysis. A correction for multitesting was applied and we had sufficient power (0.80) to detect a small-medium effect size (0.22) between two allelic frequencies each one represented by at least 300 subjects. RESULTS Intronic rs10861688 was associated with the number of depressive events corrected for the times patients were assessed during the period of observation. In particular, AA subjects (n=21) had 4.46±3.15 events, AG (n=141) had 3.08±3.17 and GG (n=342) 2.65±2.97 (p=0.0048, beta=-0.22). No other significant associations were reported. CONCLUSION We bring further evidence that genes involved in the regulation of circadian rhythms may be relevant to depressive bipolar phases. Independent confirmation analyses are mandatory.
Collapse
Affiliation(s)
- Antonio Drago
- I.R.C.C.S. "San Giovanni di Dio", Fatebenefratelli, Brescia, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Diana De Ronchi
- Department of Biomedical and Neuromotor Sciences - DIBINEM -, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences - DIBINEM -, University of Bologna, Bologna, Italy
| |
Collapse
|
27
|
Lopes-Borges J, Valvassori SS, Varela RB, Tonin PT, Vieira JS, Gonçalves CL, Streck EL, Quevedo J. Histone deacetylase inhibitors reverse manic-like behaviors and protect the rat brain from energetic metabolic alterations induced by ouabain. Pharmacol Biochem Behav 2014; 128:89-95. [PMID: 25433326 DOI: 10.1016/j.pbb.2014.11.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/03/2014] [Accepted: 11/07/2014] [Indexed: 11/19/2022]
Abstract
Studies have revealed alterations in mitochondrial complexes in the brains of bipolar patients. However, few studies have examined changes in the enzymes of the tricarboxylic acid cycle. Several preclinical studies have suggested that histone deacetylase inhibitors may have antimanic effects. The present study aims to investigate the effects of lithium, valproate and sodium butyrate, a histone deacetylase inhibitor, on the activity of tricarboxylic acid cycle enzymes in the brains of rats subjected to an animal model of mania induced by ouabain. Wistar rats received a single intracerebroventricular injection of ouabain or cerebrospinal fluid. Starting on the day following the intracerebroventricular injection, the rats were treated for 7days with intraperitoneal injections of saline, lithium, valproate or sodium butyrate. Risk-taking behavior, locomotor and exploratory activities were measured using the open-field test. Citrate synthase, succinate dehydrogenase, and malate dehydrogenase were examined in the frontal cortex and hippocampus. All treatments reversed ouabain-related risk-taking behavior and hyperactivity in the open-field test. Ouabain inhibited tricarboxylic acid cycle enzymes in the brain, and valproate and sodium butyrate but not lithium reversed this ouabain-induced dysfunction. Thus, protecting the tricarboxylic acid cycle may contribute to the therapeutic effects of histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Jéssica Lopes-Borges
- Laboratory of Neurosciences, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - Samira S Valvassori
- Laboratory of Neurosciences, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil.
| | - Roger B Varela
- Laboratory of Neurosciences, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - Paula T Tonin
- Laboratory of Neurosciences, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - Julia S Vieira
- Laboratory of Bioenergetics, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - Cinara L Gonçalves
- Laboratory of Bioenergetics, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - Emilio L Streck
- Laboratory of Bioenergetics, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil
| | - João Quevedo
- Laboratory of Neurosciences, National Institute for Translational Medicine (INCT-TM), Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC), Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC 88806-000, Brazil; Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences, University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
28
|
Curran G, Ravindran A. Lithium for bipolar disorder: a review of the recent literature. Expert Rev Neurother 2014; 14:1079-98. [DOI: 10.1586/14737175.2014.947965] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
Rybakowski JK. Response to lithium in bipolar disorder: clinical and genetic findings. ACS Chem Neurosci 2014; 5:413-21. [PMID: 24625017 DOI: 10.1021/cn5000277] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The use of lithium is a cornerstone for preventing recurrences in bipolar disorder (BD). The response of patients with bipolar disorder to lithium has different levels of magnitude. About one-third of lithium-treated patients are excellent lithium responders (ELR), showing total prevention of the episodes. A number of clinical characteristics were delineated in patients with favorable response to lithium as regards to clinical course, family history of mood disorders, and psychiatric comorbidity. We have also demonstrated that temperamental features of hypomania (a hyperthymic temperament) and a lack of cognitive disorganization predict the best results of lithium prophylaxis. A degree of prevention against manic and depressive episodes has been regarded as an endophenotype for pharmacogenetic studies. The majority of data have been gathered from so-called "candidate" gene studies. The candidates were selected on the basis of neurobiology of bipolar disorder and mechanisms of lithium action including, among others, neurotransmission, intracellular signaling, neuroprotection or circadian rhythms. We demonstrated that response to lithium has been connected with the genotype of BDNF gene and serum BDNF levels and have shown that ELR have normal cognitive functions and serum BDNF levels, even after long-term duration of the illness. A number of genome-wide association studies (GWAS) of BD have been also performed in recent years, some of which also focused on lithium response. The Consortium on Lithium Genetics (ConLiGen) has established the large sample for performing the genome-wide association study (GWAS) of lithium response in BD, and the first results have already been published.
Collapse
Affiliation(s)
- Janusz K. Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| |
Collapse
|
30
|
Rapoport SI. Lithium and the other mood stabilizers effective in bipolar disorder target the rat brain arachidonic acid cascade. ACS Chem Neurosci 2014; 5:459-67. [PMID: 24786695 DOI: 10.1021/cn500058v] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
This Review evaluates the arachidonic acid (AA, 20:4n-6) cascade hypothesis for the actions of lithium and other FDA-approved mood stabilizers in bipolar disorder (BD). The hypothesis is based on evidence in unanesthetized rats that chronically administered lithium, carbamazepine, valproate, or lamotrigine each downregulated brain AA metabolism, and it is consistent with reported upregulated AA cascade markers in post-mortem BD brain. In the rats, each mood stabilizer reduced AA turnover in brain phospholipids, cyclooxygenase-2 expression, and prostaglandin E2 concentration. Lithium and carbamazepine also reduced expression of cytosolic phospholipase A2 (cPLA2) IVA, which releases AA from membrane phospholipids, whereas valproate uncompetitively inhibited in vitro acyl-CoA synthetase-4, which recycles AA into phospholipid. Topiramate and gabapentin, proven ineffective in BD, changed rat brain AA metabolism minimally. On the other hand, the atypical antipsychotics olanzapine and clozapine, which show efficacy in BD, decreased rat brain AA metabolism by reducing plasma AA availability. Each of the four approved mood stabilizers also dampened brain AA signaling during glutamatergic NMDA and dopaminergic D2 receptor activation, while lithium enhanced the signal during cholinergic muscarinic receptor activation. In BD patients, such signaling effects might normalize the neurotransmission imbalance proposed to cause disease symptoms. Additionally, the antidepressants fluoxetine and imipramine, which tend to switch BD depression to mania, each increased AA turnover and cPLA2 IVA expression in rat brain, suggesting that brain AA metabolism is higher in BD mania than depression. The AA hypothesis for mood stabilizer action is consistent with reports that low-dose aspirin reduced morbidity in patients taking lithium, and that high n-3 and/or low n-6 polyunsaturated fatty acid diets, which in rats reduce brain AA metabolism, were effective in BD and migraine patients.
Collapse
Affiliation(s)
- Stanley I. Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
31
|
Brennand KJ, Landek-Salgado MA, Sawa A. Modeling heterogeneous patients with a clinical diagnosis of schizophrenia with induced pluripotent stem cells. Biol Psychiatry 2014; 75:936-44. [PMID: 24331955 PMCID: PMC4022707 DOI: 10.1016/j.biopsych.2013.10.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 12/28/2022]
Abstract
Schizophrenia (SZ) is a devastating complex genetic mental condition that is heterogeneous in terms of clinical etiologies, symptoms, and outcomes. Despite decades of postmortem, neuroimaging, pharmacological, and genetic studies of patients, in addition to animal models, much of the biological mechanisms that underlie the pathology of SZ remain unknown. The ability to reprogram adult somatic cells into human induced pluripotent stem cells (hiPSCs) provides a new tool that supplies live human neurons for modeling complex genetic conditions such as SZ. The purpose of this review is to discuss the technical and clinical constraints currently limiting hiPSC-based studies. We posit that reducing the clinical heterogeneity of hiPSC-based studies, by selecting subjects with common clinical manifestations or rare genetic variants, will help our ability to draw meaningful insights from the necessarily small patient cohorts that can be studied at this time.
Collapse
Affiliation(s)
- Kristen J Brennand
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | | | - Akira Sawa
- Department of Psychiatry, John Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Salloum NC, McCarthy MJ, Leckband SG, Kelsoe JR. Towards the clinical implementation of pharmacogenetics in bipolar disorder. BMC Med 2014; 12:90. [PMID: 24885933 PMCID: PMC4039055 DOI: 10.1186/1741-7015-12-90] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/29/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Bipolar disorder (BD) is a psychiatric illness defined by pathological alterations between the mood states of mania and depression, causing disability, imposing healthcare costs and elevating the risk of suicide. Although effective treatments for BD exist, variability in outcomes leads to a large number of treatment failures, typically followed by a trial and error process of medication switches that can take years. Pharmacogenetic testing (PGT), by tailoring drug choice to an individual, may personalize and expedite treatment so as to identify more rapidly medications well suited to individual BD patients. DISCUSSION A number of associations have been made in BD between medication response phenotypes and specific genetic markers. However, to date clinical adoption of PGT has been limited, often citing questions that must be answered before it can be widely utilized. These include: What are the requirements of supporting evidence? How large is a clinically relevant effect? What degree of specificity and sensitivity are required? Does a given marker influence decision making and have clinical utility? In many cases, the answers to these questions remain unknown, and ultimately, the question of whether PGT is valid and useful must be determined empirically. Towards this aim, we have reviewed the literature and selected drug-genotype associations with the strongest evidence for utility in BD. SUMMARY Based upon these findings, we propose a preliminary panel for use in PGT, and a method by which the results of a PGT panel can be integrated for clinical interpretation. Finally, we argue that based on the sufficiency of accumulated evidence, PGT implementation studies are now warranted. We propose and discuss the design for a randomized clinical trial to test the use of PGT in the treatment of BD.
Collapse
Affiliation(s)
| | | | | | - John R Kelsoe
- Department of Psychiatry (0603), University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Affiliation(s)
- Katja Dralle Mjos
- Medicinal Inorganic Chemistry Group, Department of Chemistry, The University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | | |
Collapse
|
34
|
Chen CH, Lee CS, Lee MTM, Ouyang WC, Chen CC, Chong MY, Wu JY, Tan HKL, Lee YC, Chuo LJ, Chiu NY, Tsang HY, Chang TJ, Lung FW, Chiu CH, Chang CH, Chen YS, Hou YM, Chen CC, Lai TJ, Tung CL, Chen CY, Lane HY, Su TP, Feng J, Lin JJ, Chang CJ, Teng PR, Liu CY, Chen CK, Liu IC, Chen JJ, Lu T, Fan CC, Wu CK, Li CF, Wang KHT, Wu LSH, Peng HL, Chang CP, Lu LS, Chen YT, Cheng ATA. Variant GADL1 and response to lithium therapy in bipolar I disorder. N Engl J Med 2014; 370:119-128. [PMID: 24369049 DOI: 10.1056/nejmoa1212444] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Lithium has been a first-line choice for maintenance treatment of bipolar disorders to prevent relapse of mania and depression, but many patients do not have a response to lithium treatment. METHODS We selected subgroups from a sample of 1761 patients of Han Chinese descent with bipolar I disorder who were recruited by the Taiwan Bipolar Consortium. We assessed their response to lithium treatment using the Alda scale and performed a genomewide association study on samples from one subgroup of 294 patients with bipolar I disorder who were receiving lithium treatment. We then tested the single-nucleotide polymorphisms (SNPs) that showed the strongest association with a response to lithium for association in a replication sample of 100 patients and tested them further in a follow-up sample of 24 patients. We sequenced the exons, exon-intron boundaries, and part of the promoter of the gene encoding glutamate decarboxylase-like protein 1 (GADL1) in 94 patients who had a response to lithium and in 94 patients who did not have a response in the genomewide association sample. RESULTS Two SNPs in high linkage disequilibrium, rs17026688 and rs17026651, that are located in the introns of GADL1 showed the strongest associations in the genomewide association study (P=5.50×10(-37) and P=2.52×10(-37), respectively) and in the replication sample of 100 patients (P=9.19×10(-15) for each SNP). These two SNPs had a sensitivity of 93% for predicting a response to lithium and differentiated between patients with a good response and those with a poor response in the follow-up cohort. Resequencing of GADL1 revealed a novel variant, IVS8+48delG, which lies in intron 8 of the gene, is in complete linkage disequilibrium with rs17026688 and is predicted to affect splicing. CONCLUSIONS Genetic variations in GADL1 are associated with the response to lithium maintenance treatment for bipolar I disorder in patients of Han Chinese descent. (Funded by Academia Sinica and others.).
Collapse
|
35
|
Tobe BTD, Brandel MG, Nye JS, Snyder EY. Implications and limitations of cellular reprogramming for psychiatric drug development. Exp Mol Med 2013; 45:e59. [PMID: 24232258 PMCID: PMC3849573 DOI: 10.1038/emm.2013.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 08/28/2013] [Indexed: 12/28/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) derived from somatic cells of patients have opened possibilities for in vitro modeling of the physiology of neural (and other) cells in psychiatric disease states. Issues in early stages of technology development include (1) establishing a library of cells from adequately phenotyped patients, (2) streamlining laborious, costly hiPSC derivation and characterization, (3) assessing whether mutations or other alterations introduced by reprogramming confound interpretation, (4) developing efficient differentiation strategies to relevant cell types, (5) identifying discernible cellular phenotypes meaningful for cyclic, stress induced or relapsing-remitting diseases, (6) converting phenotypes to screening assays suitable for genome-wide mechanistic studies or large collection compound testing and (7) controlling for variability in relation to disease specificity amidst low sample numbers. Coordination of material for reprogramming from patients well-characterized clinically, genetically and with neuroimaging are beginning, and initial studies have begun to identify cellular phenotypes. Finally, several psychiatric drugs have been found to alter reprogramming efficiency in vitro, suggesting further complexity in applying hiPSCs to psychiatric diseases or that some drugs influence neural differentiation moreso than generally recognized. Despite these challenges, studies utilizing hiPSCs may eventually serve to fill essential niches in the translational pipeline for the discovery of new therapeutics.
Collapse
Affiliation(s)
- Brian T D Tobe
- 1] Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, Burnham Institute for Medical Research, La Jolla, CA, USA [2] Department of Psychiatry, Veterans Administration Medical Center, La Jolla, CA, USA
| | | | | | | |
Collapse
|
36
|
Ehret MJ, Baker W, O'Neill H. BDNF Val66Met polymorphism and lithium response: a meta-analysis. Per Med 2013; 10:777-784. [PMID: 29776284 DOI: 10.2217/pme.13.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIM To characterize the impact of the Val66Met polymorphism on lithium response in patients with bipolar disorder. METHODS A systematic literature search of MEDLINE, Web of Science, PsychINFO and Cochrane CENTRAL was conducted from the earliest possible date through to 1 July 2012. The search was performed using the following medical subject headings: bipolar disorder, lithium, lithium carbonate, pharmacogenomics, pharmacogenetics, polymorphism and brain-derived neurotrophic factor. Five of 71 identified studies met the inclusion criteria. Data were abstracted using a standardized data abstraction tool. For categorical end points, the pooled odds ratio with 95% CI was calculated. Random effects models were used for analysis. RESULTS The Val66Met polymorphism did not predict response to prophylactic lithium in this combined population (odds ratio: 2.67; p = 0.078). CONCLUSION This analysis suggests that the Val66Met polymorphism does not predict response to lithium treatment in bipolar disorder in this combined population. Prospective studies are needed to clearly define the role of Val66Met polymorphism of BDNF in lithium response.
Collapse
Affiliation(s)
- Megan J Ehret
- Department of Pharmacy Practice, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269, USA.
| | - William Baker
- Department of Pharmacy Practice, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269, USA
| | - Hannah O'Neill
- Department of Pharmacy Practice, School of Pharmacy, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, CT 06269, USA
| |
Collapse
|
37
|
McCarthy MJ, Wei H, Marnoy Z, Darvish RM, McPhie DL, Cohen BM, Welsh DK. Genetic and clinical factors predict lithium's effects on PER2 gene expression rhythms in cells from bipolar disorder patients. Transl Psychiatry 2013; 3:e318. [PMID: 24150227 PMCID: PMC3818008 DOI: 10.1038/tp.2013.90] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 09/08/2013] [Indexed: 11/09/2022] Open
Abstract
Bipolar disorder (BD) is associated with abnormal circadian rhythms. In treatment responsive BD patients, lithium (Li) stabilizes mood and reduces suicide risk. Li also affects circadian rhythms and expression of 'clock genes' that control them. However, the extent to which BD, Li and the circadian clock share common biological mechanisms is unknown, and there have been few direct measurements of clock gene function in samples from BD patients. Hence, the role of clock genes in BD and Li treatment remains unclear. Skin fibroblasts from BD patients (N=19) or healthy controls (N=19) were transduced with Per2::luc, a rhythmically expressed, bioluminescent circadian clock reporter gene, and rhythms were measured for 5 consecutive days. Rhythm amplitude and period were compared between BD cases and controls with and without Li. Baseline period was longer in BD cases than in controls. Li 1 mM increased amplitude in controls by 36%, but failed to do so in BD cases. Li 10 mM lengthened period in both BD cases and controls. Analysis of clock gene variants revealed that PER3 and RORA genotype predicted period lengthening by Li, whereas GSK3β genotype predicted rhythm effects of Li, specifically among BD cases. Analysis of BD cases by clinical history revealed that cells from past suicide attempters were more likely to show period lengthening with Li 1 mM. Finally, Li enhanced the resynchronization of damped rhythms, suggesting a mechanism by which Li could act therapeutically in BD. Our work suggests that the circadian clock's response to Li may be relevant to molecular pathology of BD.
Collapse
Affiliation(s)
- M J McCarthy
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, University of California, San Diego, CA, USA,Department of Psychiatry, University of California, San Diego, CA, USA,Center for Chronobiology, University of California, San Diego, CA, USA,Psychiatry Service, Veterans Affairs San Diego Healthcare System, University of California, 3350 La Jolla Village Drive, MC 116A, San Diego, 92161 CA, USA. E-mail:
| | - H Wei
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, University of California, San Diego, CA, USA,Department of Psychiatry, University of California, San Diego, CA, USA,Center for Chronobiology, University of California, San Diego, CA, USA
| | - Z Marnoy
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, University of California, San Diego, CA, USA,Department of Psychiatry, University of California, San Diego, CA, USA,Center for Chronobiology, University of California, San Diego, CA, USA
| | - R M Darvish
- Department of Psychiatry, University of California, San Diego, CA, USA,Center for Chronobiology, University of California, San Diego, CA, USA
| | - D L McPhie
- Harvard McLean Hospital, Belmont, MA, USA
| | - B M Cohen
- Harvard McLean Hospital, Belmont, MA, USA
| | - D K Welsh
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, University of California, San Diego, CA, USA,Department of Psychiatry, University of California, San Diego, CA, USA,Center for Chronobiology, University of California, San Diego, CA, USA
| |
Collapse
|
38
|
Abstract
Pharmacogenetics brought the promise of matching individuals with treatments that would be efficacious while minimizing adverse events. This has been long needed in psychiatry, where treatment options have been empirical and treatment choices have been made largely based on clinical judgment. The efficacy and tolerability of antidepressants, the most common drugs used in mood disorders, have been widely studied in pharmacogenetics. Genetic association studies have been reported for pharmacokinetic genes such as the CYP450 isoenzymes or MDR1, and pharmacodynamic genes such as the serotonin transporter (SLC6A4) or the serotonin 2A receptor (HTR2A). However, despite the large number of reports, clinically useful predictors are still scarce for antidepressant monotherapy. Pharmacogenetic predictors of efficacy for mood stabilizers such as lithium and anticonvulsants have not had a dissimilar fate, and clinically meaningful markers are yet to emerge. The lack of consistent results may be in part due to small samples of heterogeneous populations and lack of consensus on phenotype definitions. Current pharmacogenetic recommendations include testing for HLA-B*1502 when using carbamazepine in Asian ancestry populations to prevent Stevens–Johnson syndrome, CYP2D6 genotypes when using pimozide, and CYP2D6 in polypharmacy to minimize drug interactions. This review, which is aimed at clinicians, lays the basis for understanding strengths and weaknesses of pharmacogenetic studies and outlines current clinical uses of these biomarkers.
Collapse
|
39
|
Differential antidepressant-like response to lithium treatment between mouse strains: effects of sex, maternal care, and mixed genetic background. Psychopharmacology (Berl) 2013; 228:411-8. [PMID: 23503701 PMCID: PMC3707960 DOI: 10.1007/s00213-013-3045-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 02/23/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND Lithium is a mood stabilizer with both antidepressant and antimanic properties, however its mechanism of action is unclear. Identifying the genetic factors that influence lithium's therapeutic actions will be an important step to assist in identifying such mechanisms. We previously reported that lithium treatment of male mice has antidepressant-like effects in the C57BL/6J strain but that such effects were absent in the BALB/cJ strain. OBJECTIVES This study aimed to assess the roles of both genetic and non-genetic factors such as sex and non-shared environmental conditions that may mediate differential behavioral responses to lithium. METHODS Mice were treated with lithium for 10 days and then tested in the forced swim test followed by lithium discontinuation and retesting to assess effects of lithium withdrawal. We also assessed effects of sex and cross-fostering on lithium response between the C57BL/6J and BALB/cJ strains, and antidepressant-like effects of lithium in the hybrid CB6F1/J strain that is derived from C57BL/6J and BALB/cJ parental strains. RESULTS Neither sex nor maternal care significantly influenced the differential antidepressant-like response to lithium. Withdrawal from lithium treatment reversed antidepressant-like effects in the C57BL/6J strain but had no effects in BALB/cJ mice. Lithium treatment did not result in antidepressant-like effects in the CB6F1/J strain. CONCLUSIONS Genetic factors are likely primarily responsible for differential antidepressant-like effects of lithium in the C57BL/6J and BALB/cJ strains. Future studies identifying such genetic factors may help to elucidate the neurobiological mechanisms of lithium's therapeutic actions.
Collapse
|
40
|
Manchia M, Adli M, Akula N, Ardau R, Aubry JM, Backlund L, Banzato CEM, Baune BT, Bellivier F, Bengesser S, Biernacka JM, Brichant-Petitjean C, Bui E, Calkin CV, Cheng ATA, Chillotti C, Cichon S, Clark S, Czerski PM, Dantas C, Zompo MD, DePaulo JR, Detera-Wadleigh SD, Etain B, Falkai P, Frisén L, Frye MA, Fullerton J, Gard S, Garnham J, Goes FS, Grof P, Gruber O, Hashimoto R, Hauser J, Heilbronner U, Hoban R, Hou L, Jamain S, Kahn JP, Kassem L, Kato T, Kelsoe JR, Kittel-Schneider S, Kliwicki S, Kuo PH, Kusumi I, Laje G, Lavebratt C, Leboyer M, Leckband SG, López Jaramillo CA, Maj M, Malafosse A, Martinsson L, Masui T, Mitchell PB, Mondimore F, Monteleone P, Nallet A, Neuner M, Novák T, O’Donovan C, Ösby U, Ozaki N, Perlis RH, Pfennig A, Potash JB, Reich-Erkelenz D, Reif A, Reininghaus E, Richardson S, Rouleau GA, Rybakowski JK, Schalling M, Schofield PR, Schubert OK, Schweizer B, Seemüller F, Grigoroiu-Serbanescu M, Severino G, Seymour LR, Slaney C, Smoller JW, Squassina A, Stamm T, Steele J, Stopkova P, Tighe SK, Tortorella A, Turecki G, Wray NR, Wright A, Zandi PP, Zilles D, Bauer M, Rietschel M, McMahon FJ, Schulze TG, Alda M. Assessment of Response to Lithium Maintenance Treatment in Bipolar Disorder: A Consortium on Lithium Genetics (ConLiGen) Report. PLoS One 2013; 8:e65636. [PMID: 23840348 PMCID: PMC3686769 DOI: 10.1371/journal.pone.0065636] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/26/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The assessment of response to lithium maintenance treatment in bipolar disorder (BD) is complicated by variable length of treatment, unpredictable clinical course, and often inconsistent compliance. Prospective and retrospective methods of assessment of lithium response have been proposed in the literature. In this study we report the key phenotypic measures of the "Retrospective Criteria of Long-Term Treatment Response in Research Subjects with Bipolar Disorder" scale currently used in the Consortium on Lithium Genetics (ConLiGen) study. MATERIALS AND METHODS Twenty-nine ConLiGen sites took part in a two-stage case-vignette rating procedure to examine inter-rater agreement [Kappa (κ)] and reliability [intra-class correlation coefficient (ICC)] of lithium response. Annotated first-round vignettes and rating guidelines were circulated to expert research clinicians for training purposes between the two stages. Further, we analyzed the distributional properties of the treatment response scores available for 1,308 patients using mixture modeling. RESULTS Substantial and moderate agreement was shown across sites in the first and second sets of vignettes (κ = 0.66 and κ = 0.54, respectively), without significant improvement from training. However, definition of response using the A score as a quantitative trait and selecting cases with B criteria of 4 or less showed an improvement between the two stages (ICC1 = 0.71 and ICC2 = 0.75, respectively). Mixture modeling of score distribution indicated three subpopulations (full responders, partial responders, non responders). CONCLUSIONS We identified two definitions of lithium response, one dichotomous and the other continuous, with moderate to substantial inter-rater agreement and reliability. Accurate phenotypic measurement of lithium response is crucial for the ongoing ConLiGen pharmacogenomic study.
Collapse
Affiliation(s)
- Mirko Manchia
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Nirmala Akula
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University-Hospital of Cagliari, Cagliari, Italy
| | - Jean-Michel Aubry
- Hôpitaux Universitaires de Genève, Department of Mental Health and Psychiatry, Geneva, Switzerland
| | - Lena Backlund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Bernhard T. Baune
- Department of Psychiatry, The University of Adelaide, Adelaide, Australia
| | - Frank Bellivier
- Assistance publique - Hôpitaux de Paris, Groupe Hospitalier Lariboisière-F. Widal, Pôle de Psychiatrie, Paris, France
| | | | - Joanna M. Biernacka
- Department of Psychiatry, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Clara Brichant-Petitjean
- Assistance publique - Hôpitaux de Paris, Groupe Hospitalier Lariboisière-F. Widal, Pôle de Psychiatrie, Paris, France
| | - Elise Bui
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Cynthia V. Calkin
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Tai Ann Cheng
- Division of Epidemiology and Genetics, Academia Sinica, Institute of Biomedical Sciences, Taipei, Taiwan
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University-Hospital of Cagliari, Cagliari, Italy
| | - Sven Cichon
- Department of Genomics, Life and Brain Center and Institute of Human Genetics, Bonn University, Bonn, Germany
| | - Scott Clark
- Department of Psychiatry, The University of Adelaide, Adelaide, Australia
| | - Piotr M. Czerski
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Clarissa Dantas
- Department of Psychiatry, University of Campinas, Campinas, Brazil
| | - Maria Del Zompo
- Unit of Clinical Pharmacology, University-Hospital of Cagliari, Cagliari, Italy
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, Cagliari, Italy
| | - J. Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sevilla D. Detera-Wadleigh
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Bruno Etain
- Institut National de la Santé et de la Recherche Médicale, Unité 955, Institut Mondor de Recherche Biomédicale, Equipe 15, Faculté de médecine, Créteil, France
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Louise Frisén
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mark A. Frye
- Department of Psychiatry, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jan Fullerton
- Neuroscience Research Australia - Genetics of Mental Illness and Brain Function, Sydney, Australia
| | - Sébastien Gard
- Service de psychiatrie, Hôpital Charles Perrens, Bordeaux, France
| | - Julie Garnham
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Fernando S. Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Paul Grof
- Mood Disorders Center of Ottawa, Ottawa, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Oliver Gruber
- Department of Psychiatry and Psychotherapy, Georg-August-Universität, Göttingen, Germany
| | | | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Urs Heilbronner
- Department of Psychiatry and Psychotherapy, Georg-August-Universität, Göttingen, Germany
| | - Rebecca Hoban
- Department of Psychiatry, University of California San Diego, San Diego, California, United States of America
- Department of Psychiatry, Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
| | - Liping Hou
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Stéphane Jamain
- Institut National de la Santé et de la Recherche Médicale, Unité 955, Institut Mondor de Recherche Biomédicale, Equipe 15, Faculté de médecine, Créteil, France
| | - Jean-Pierre Kahn
- Service de Psychiatrie et Psychologie Clinique, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Layla Kassem
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Saitama, Japan
| | - John R. Kelsoe
- Department of Psychiatry, University of California San Diego, San Diego, California, United States of America
- Department of Psychiatry, Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Sebastian Kliwicki
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Ichiro Kusumi
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Saitama, Japan
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Gonzalo Laje
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Marion Leboyer
- Institut National de la Santé et de la Recherche Médicale, Unité 955, Institut Mondor de Recherche Biomédicale, Equipe 15, Faculté de médecine, Créteil, France
| | - Susan G. Leckband
- Department of Psychiatry, University of California San Diego, San Diego, California, United States of America
- Department of Pharmacy, Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, California, United States of America
| | | | - Mario Maj
- Department of Psychiatry, University of Napoli, Napoli, Italy
| | - Alain Malafosse
- Hôpitaux Universitaires de Genève, Department of Mental Health and Psychiatry, Geneva, Switzerland
| | - Lina Martinsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Takuya Masui
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Philip B. Mitchell
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, Australia
| | - Frank Mondimore
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | - Audrey Nallet
- Hôpitaux Universitaires de Genève, Department of Mental Health and Psychiatry, Geneva, Switzerland
| | - Maria Neuner
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Tomás Novák
- Prague Psychiatric Center, University of Prague, Prague, Czech Republic
| | - Claire O’Donovan
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Urban Ösby
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Norio Ozaki
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrea Pfennig
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Germany
| | - James B. Potash
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, United States of America
| | - Daniela Reich-Erkelenz
- Department of Psychiatry and Psychotherapy, Georg-August-Universität, Göttingen, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Eva Reininghaus
- Department of Psychiatry, Medical University of Graz, Graz, Austria
| | - Sara Richardson
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Guy A. Rouleau
- Centre of Excellence in Neuroscience of Université de Montréal, Centre Hospitalier de l’Université de Montréal and Department of Medicine, Université de Montréal, Montréal, Canada
| | - Janusz K. Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter R. Schofield
- Neuroscience Research Australia - Genetics of Mental Illness and Brain Function, Sydney, Australia
| | - Oliver K. Schubert
- Department of Psychiatry, The University of Adelaide, Adelaide, Australia
| | - Barbara Schweizer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Florian Seemüller
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Maria Grigoroiu-Serbanescu
- Alexandru Obregia Psychiatric Hospital, Biometric Psychiatric Genetics Research Unit, Bucharest, Romania
| | - Giovanni Severino
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, Cagliari, Italy
| | - Lisa R. Seymour
- Department of Psychiatry, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Claire Slaney
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alessio Squassina
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, Cagliari, Italy
| | - Thomas Stamm
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Berlin, Germany
| | - Jo Steele
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Pavla Stopkova
- Prague Psychiatric Center, University of Prague, Prague, Czech Republic
| | - Sarah K. Tighe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montréal, Canada
| | - Naomi R. Wray
- The University of Queensland, Queensland Brain Institute, Brisbane, Australia
| | - Adam Wright
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, Australia
| | - Peter P. Zandi
- Johns Hopkins Bloomberg School of Public Health, Department of Mental Health, Baltimore, Maryland, United States of America
| | - David Zilles
- Department of Psychiatry and Psychotherapy, Georg-August-Universität, Göttingen, Germany
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Francis J. McMahon
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Thomas G. Schulze
- Human Genetics Branch, Division of Intramural Research Programs, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Psychiatry and Psychotherapy, Georg-August-Universität, Göttingen, Germany
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
- * E-mail: (TS); (M. Alda)
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail: (TS); (M. Alda)
| |
Collapse
|
41
|
Abstract
Suicide and bipolar disorder (BD) are challenging, complex, and intertwined areas of study in contemporary psychiatry. Indeed, BD is associated with the highest lifetime risk for suicide attempt and completion of all the psychiatric conditions. Given that several clinical risk factors for both suicide and BD have been well noted in the literature, exploring the neurobiological aspects of suicide in BD may provide insights into both preventive measures and future novel treatments. This review synthesizes findings regarding the neurobiological aspects of suicide and, when applicable, their link to BD. Neurochemical findings, genes/epigenetics, and potential molecular targets for current or future treatments are discussed. The role of endophenotypes and related proximal and distal risk factors underlying suicidal behavior are also explored. Lastly, we discuss the manner in which preclinical work on aggression and impulsivity may provide additional insights for the future development of novel treatments.
Collapse
|
42
|
Abstract
Mood stabilizers form a cornerstone in the long-term treatment of bipolar disorder. The first representative of their family was lithium, still considered a prototype drug for the prevention of manic and depressive recurrences in bipolar disorder. Along with carbamazepine and valproates, lithium belongs to the first generation of mood stabilizers, which appeared in psychiatric treatment in the 1960s. Atypical antipsychotics with mood-stabilizing properties and lamotrigine, which were introduced in the mid-1990 s, form the second generation of such drugs. The response of patients with bipolar disorder to mood stabilizers has different levels of magnitude. About one-third of lithium-treated patients are excellent responders, showing total prevention of the episodes, and these patients are clinically characterized by an episodic clinical course, complete remission, a bipolar family history, low psychiatric co-morbidity and a hyperthymic temperament. It has been suggested that responders to carbamazepine or lamotrigine may differ clinically from responders to lithium. The main phenotype of the response to mood stabilizers is a degree of prevention against recurrences of manic and depressive episodes during long-term treatment. The most specific scale in this respect is the so-called Alda scale, where retrospective assessment of lithium response is scored on a 0-10 scale. The vast majority of data on genetic influences on the response to mood stabilizers has been gathered in relation to lithium. The studies on the mechanisms of action of lithium and on the neurobiology of bipolar disorder have led to the identification of a number of candidate genes. The genes studied for their association with lithium response have been those connected with neurotransmitters (serotonin, dopamine and glutamate), second messengers (phosphatidyl inositol [PI], cyclic adenosine-monophosphate [cAMP] and protein kinase C [PKC] pathways), substances involved in neuroprotection (brain-derived neurotrophic factor [BDNF] and glycogen synthase kinase 3-β [GSK-3β]) and a number of other miscellaneous genes. There are no published pharmacogenomic studies of mood stabilizers other than lithium, except for one study of the X-box binding protein 1 (XBP1) gene in relation to the efficacy of valproate. In recent years, a number of genome-wide association studies (GWAS) in bipolar disorders have been performed and some of those have also focused on lithium response. They suggest roles for the glutamatergic receptor AMPA (GRIA2) gene and the amiloride-sensitive cation channel 1 neuronal (ACCN1) gene in long-term lithium response. A promise for better elucidating the genetics of lithium response has been created by the formation of the Consortium on Lithium Genetics (ConLiGen) to establish the largest sample, to date, for the GWAS of lithium response in bipolar disorder. The sample currently comprises more than 1,200 patients, characterized by their response to lithium treatment according to the Alda scale. Preliminary results from this international study suggest a possible involvement of the sodium bicarbonate transporter (SLC4A10) gene in lithium response. It is concluded that the pharmacogenetics of response to mood stabilizers has recently become a growing field of research, especially so far as the pharmacogenetics of the response to lithium is concerned. Clearly, the ConLiGen project is a highly significant step in this research. Although the results of pharmacogenetic studies are of significant scientific value, their possible practical implications are yet to be seen.
Collapse
|
43
|
Wall CA, Croarkin PE, Swintak C, Koplin BA. Psychiatric pharmacogenomics in pediatric psychopharmacology. Child Adolesc Psychiatr Clin N Am 2012; 21:773-88. [PMID: 23040901 DOI: 10.1016/j.chc.2012.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This article provides an overview of where psychiatric pharmacogenomic testing stands as an emerging clinical tool in modern psychotropic prescribing practice, specifically in the pediatric population. This practical discussion is organized around the state of psychiatric pharmacogenomics research when choosing psychopharmacologic interventions in the most commonly encountered mental illnesses in youth. As with the rest of the topics on psychopharmacology for children and adolescents in this publication, a clinical vignette is presented, this one highlighting a clinical case of a 16 year old genotyped during hospitalization for recalcitrant depression.
Collapse
Affiliation(s)
- Christopher A Wall
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
44
|
Ramadan E, Basselin M, Chang L, Chen M, Ma K, Rapoport SI. Chronic lithium feeding reduces upregulated brain arachidonic acid metabolism in HIV-1 transgenic rat. J Neuroimmune Pharmacol 2012; 7:701-13. [PMID: 22760927 PMCID: PMC3478068 DOI: 10.1007/s11481-012-9381-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/04/2012] [Indexed: 01/09/2023]
Abstract
HIV-1 transgenic (Tg) rats, a model for human HIV-1 associated neurocognitive disorder (HAND), show upregulated markers of brain arachidonic acid (AA) metabolism with neuroinflammation after 7 months of age. Since lithium decreases AA metabolism in a rat lipopolysaccharide model of neuroinflammation, and may be useful in HAND, we hypothesized that lithium would dampen upregulated brain AA metabolism in HIV-1 Tg rats. Regional brain AA incorporation coefficients k* and rates J ( in ), markers of AA signaling and metabolism, were measured in 81 brain regions using quantitative autoradiography, after intravenous [1-(14) C]AA infusion in unanesthetized 10-month-old HIV-1 Tg and age-matched wildtype rats that had been fed a control or LiCl diet for 6 weeks. k* and J ( in ) for AA were significantly higher in HIV-1 Tg than wildtype rats fed the control diet. Lithium feeding reduced plasma unesterified AA concentration in both groups and J ( in ) in wildtype rats, and blocked increments in k* (19 of 54 regions) and J ( in ) (77 of 81 regions) in HIV-1 Tg rats. These in vivo neuroimaging data indicate that lithium treatment dampened upregulated brain AA metabolism in HIV-1 Tg rats. Lithium may improve cognitive dysfunction and be neuroprotective in HIV-1 patients with HAND through a comparable effect.
Collapse
Affiliation(s)
- Epolia Ramadan
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Mireille Basselin
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa Chang
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Mei Chen
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Kaizong Ma
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Stanley I. Rapoport
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
45
|
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
|
46
|
Leckband SG, McCarthy M, Kelsoe JR. The pharmacogenomics of mood stabilizer response in bipolar disorder. Ment Health Clin 2012. [DOI: 10.9740/mhc.n99509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bipolar Disorder (BD) is a common psychiatric illness that has a recurring course and 17% - 24% lifetime prevalence of suicide attempts. Difficulty in diagnosis and individual variation in response point to a great need for personalized medicine. Studies to date have focused largely on lithium and suggest the idea that different medication responses may reflect different disease mechanisms. An overview of current pharmacogenomic studies will be presented and needs and future directions discussed.
Collapse
Affiliation(s)
- Susan G. Leckband
- Veterans Affairs San Diego Healthcare System
- University of California, San Diego
| | - Michael McCarthy
- Veterans Affairs San Diego Healthcare System
- University of California, San Diego
| | - John R. Kelsoe
- Veterans Affairs San Diego Healthcare System
- University of California, San Diego
| |
Collapse
|
47
|
A survey of genomic studies supports association of circadian clock genes with bipolar disorder spectrum illnesses and lithium response. PLoS One 2012; 7:e32091. [PMID: 22384149 PMCID: PMC3285204 DOI: 10.1371/journal.pone.0032091] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 01/23/2012] [Indexed: 11/19/2022] Open
Abstract
Circadian rhythm abnormalities in bipolar disorder (BD) have led to a search for genetic abnormalities in circadian “clock genes” associated with BD. However, no significant clock gene findings have emerged from genome-wide association studies (GWAS). At least three factors could account for this discrepancy: complex traits are polygenic, the organization of the clock is more complex than previously recognized, and/or genetic risk for BD may be shared across multiple illnesses. To investigate these issues, we considered the clock gene network at three levels: essential “core” clock genes, upstream circadian clock modulators, and downstream clock controlled genes. Using relaxed thresholds for GWAS statistical significance, we determined the rates of clock vs. control genetic associations with BD, and four additional illnesses that share clinical features and/or genetic risk with BD (major depression, schizophrenia, attention deficit/hyperactivity). Then we compared the results to a set of lithium-responsive genes. Associations with BD-spectrum illnesses and lithium-responsiveness were both enriched among core clock genes but not among upstream clock modulators. Associations with BD-spectrum illnesses and lithium-responsiveness were also enriched among pervasively rhythmic clock-controlled genes but not among genes that were less pervasively rhythmic or non-rhythmic. Our analysis reveals previously unrecognized associations between clock genes and BD-spectrum illnesses, partly reconciling previously discordant results from past GWAS and candidate gene studies.
Collapse
|
48
|
Abstract
As shown by clinical genetic studies, affective and anxiety disorders are complex genetic disorders with genetic and environmental factors interactively determining their respective pathomechanism. Advances in molecular genetic techniques including linkage studies, association studies, and genome-wide association studies allow for the detailed dissection of the genetic influence on the development of these disorders. Besides the molecular genetic investigation of categorical entities according to standardized diagnostic criteria, intermediate phenotypes comprising neurobiological or neuropsychological traits (e.g., neuronal correlates of emotional processing) that are linked to the disease of interest and that are heritable, have been proposed to be closer to the underlying genotype than the overall disease phenotype. These intermediate phenotypes are dimensional and more precisely defined than the categorical disease phenotype, and therefore have attracted much interest in the genetic investigation of affective and anxiety disorders. Given the complex genetic nature of affective and anxiety disorders with an interaction of multiple risk genes and environmental influences, the interplay of genetic factors with environmental factors is investigated by means of gene-environment interaction (GxE) studies. Pharmacogenetic studies aid in the dissection of the genetically influenced heterogeneity of psychotropic drug response and may contribute to the development of a more individualized treatment of affective and anxiety disorders. Finally, there is some evidence for genetic factors potentially shared between affective and anxiety disorders pointing to a possible overlapping phenotype between anxiety disorders and depression.
Collapse
Affiliation(s)
- Katharina Domschke
- Department of Psychiatry, University of Würzburg, Füchsleinstrasse 15, D-97080, Würzburg, Germany,
| | | |
Collapse
|
49
|
Can A, Blackwell RA, Piantadosi SC, Dao DT, O’Donnell KC, Gould TD. Antidepressant-like responses to lithium in genetically diverse mouse strains. GENES, BRAIN, AND BEHAVIOR 2011; 10:434-43. [PMID: 21306560 PMCID: PMC3107888 DOI: 10.1111/j.1601-183x.2011.00682.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A mood stabilizing and antidepressant response to lithium is only found in a subgroup of patients with bipolar disorder and depression. Identifying strains of mice that manifest differential behavioral responses to lithium may assist in the identification of genomic and other biologic factors that play a role in lithium responsiveness. Mouse strains were tested in the forced swim test (FST), tail suspension test (TST) and open-field test after acute and chronic systemic and intracerebroventricular (ICV) lithium treatments. Serum and brain lithium levels were measured. Three (129S6/SvEvTac, C3H/HeNHsd and C57BL/6J) of the eight inbred strains tested, and one (CD-1) of the three outbred strains, showed an antidepressant-like response in the FST following acute systemic administration of lithium. The three responsive inbred strains, as well as the DBA/2J strain, displayed antidepressant-like responses to lithium in the FST after chronic administration of lithium. However, in the TST, acute lithium resulted in an antidepressant-like effect only in C3H/HeNHsd mice. Only C57BL/6J and DBA/2J showed an antidepressant-like response to lithium in the TST after chronic administration. ICV lithium administration resulted in a similar response profile in BALB/cJ (non-responsive) and C57BL/6J (responsive) strains. Serum and brain lithium concentrations showed that behavioral results were not because of differential pharmacokinetics of lithium in individual strains, suggesting that genetic factors likely regulate these behavioral responses to lithium. Our results indicate that antidepressant-like responses to lithium in tests of antidepressant efficacy varies among genetically diverse mouse strains. These results will assist in identifying genomic factors associated with lithium responsiveness and the mechanisms of lithium action.
Collapse
Affiliation(s)
- Adem Can
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD
| | - Robert A. Blackwell
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD
| | - Sean C. Piantadosi
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD
| | - David T. Dao
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD
| | | | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore MD
| |
Collapse
|