1
|
Bai Z, Li P, Gao X, Zu G, Jiang A, Wu K, Mechawar N, Turecki G, Lehnert K, Snell RG, Zhou J, Hu J, Yan B, Chen L, Li W, Chen Y, Liu S, Zhu Y, You L. Exploring PDE5A upregulation in bipolar disorder: insights from single-nucleus RNA sequencing of human basal ganglia. Transl Psychiatry 2024; 14:494. [PMID: 39695100 DOI: 10.1038/s41398-024-03202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
Basal ganglia is proposed to mediate symptoms underlying bipolar disorder (BD). To understand the cell type-specific gene expression and network changes of BD basal ganglia, we performed single-nucleus RNA sequencing of 30,752 nuclei from caudate, putamen, globus pallidus, and substantia nigra of control human postmortem brain and 24,672 nuclei from BD brain. Differential expression analysis revealed major difference lying in caudate, with BD medium spiny neurons (MSNs) expressing significantly higher PDE5A, a cGMP-specific phosphodiesterase. Gene co-expression analysis (WGCNA) showed a strong correlation of caudate MSNs and gene module green, with a PDE5A-containing hub gene network. Gene regulatory network analysis (SCENIC) indicated key regulons among different cell types and basal ganglia regions, with downstream targets of key transcriptional factors showing overlapping genes such as PDEs. Upregulation of PDE5A was further validated in 7 pairs of control and BD caudate sections. Overexpression of PDE5A in primary cultured lateral ganglion eminence-derived striatal neurons led to decreased dendrite complexity, increased apoptosis, and enhanced neuronal excitability and membrane resistance. This effect could be rescued by PDE5 specific inhibitor, tadalafil. Overexpression of PDE5A in mouse striatum by stereotaxic injection caused a decreased cGMP level, an increased gene expression profile of neuroinflammation, and BD-like behaviors. Collectively, our findings provided cell type-specific gene expression profile, and indicated a causative role of PDE5A upregulation in BD basal ganglia. This study provides a single-nucleus transcriptomic profile of human control and bipolar disorder (BD) basal ganglia. Differential expression, gene co-expression, and gene regulatory network analyses collectively indicated upregulation of PDE5A in BD caudate medium spiny neurons (MSNs), which was further validated in another cohort of BD brains. The causative role of PDE5A upregulation in BD etiology is supported by the effects of PDE5A overexpression in cultured mouse MSNs in vitro and in adult mouse striatum in vivo. The former led to reduced dendrite complexity, increased apoptosis, and neuronal hyper-excitability, which could be rescued by PDE5 specific inhibitor tadalafil. The latter caused lower cGMP levels, upregulated genes associated with neuroinflammation, and BD-like behaviors.
Collapse
Affiliation(s)
- Zhixin Bai
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Peilong Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xu Gao
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
- Shanghai Changning Mental Health Center, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China
| | - Gaoyu Zu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Andrew Jiang
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Keting Wu
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Klaus Lehnert
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Russell G Snell
- Applied Translational Genetics Group, School of Biological Sciences, the University of Auckland, Auckland, New Zealand
| | - Jin Zhou
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jia Hu
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Bingbing Yan
- Neo-Biotechnology Limited Company, Shanghai, China
| | - Liang Chen
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wensheng Li
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - You Chen
- Shanghai Yangpu District Mental Health Center, Mental Health Center Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China.
| | - Shuai Liu
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China.
- Shanghai Changning Mental Health Center, Shanghai, China.
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China.
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Zhang Y, Wu J, Zheng Y, Xu Y, Yu Z, Ping Y. Voltage Gated Ion Channels and Sleep. J Membr Biol 2024; 257:269-280. [PMID: 39354150 DOI: 10.1007/s00232-024-00325-0] [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: 07/31/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Ion channels are integral components of the nervous system, playing a pivotal role in shaping membrane potential, neuronal excitability, synaptic transmission and plasticity. Dysfunction in these channels, such as improper expression or localization, can lead to irregular neuronal excitability and synaptic communication, which may manifest as various behavioral abnormalities, including disrupted rest-activity cycles. Research has highlighted the significant impact of voltage gated ion channels on sleep parameters, influencing sleep latency, duration and waveforms. Furthermore, these ion channels have been implicated in the vulnerability to, and the pathogenesis of, several neurological and psychiatric disorders, including epilepsy, autism, schizophrenia, and Alzheimer's disease (AD). In this comprehensive review, we aim to provide a summary of the regulatory role of three predominant types of voltage-gated ion channels-calcium (Ca2+), sodium (Na+), and potassium (K+)-in sleep across species, from flies to mammals. We will also discuss the association of sleep disorders with various human diseases that may arise from the dysfunction of these ion channels, thereby underscoring the potential therapeutic benefits of targeting specific ion channel subtypes for sleep disturbance treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiawen Wu
- Faculty of Brain Sciences, University College London, London, UK
| | - Yuxian Zheng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yangkun Xu
- Tandon School of Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Ziqi Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
3
|
Mangione W, Falls Z, Samudrala R. Effective holistic characterization of small molecule effects using heterogeneous biological networks. Front Pharmacol 2023; 14:1113007. [PMID: 37180722 PMCID: PMC10169664 DOI: 10.3389/fphar.2023.1113007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
The two most common reasons for attrition in therapeutic clinical trials are efficacy and safety. We integrated heterogeneous data to create a human interactome network to comprehensively describe drug behavior in biological systems, with the goal of accurate therapeutic candidate generation. The Computational Analysis of Novel Drug Opportunities (CANDO) platform for shotgun multiscale therapeutic discovery, repurposing, and design was enhanced by integrating drug side effects, protein pathways, protein-protein interactions, protein-disease associations, and the Gene Ontology, and complemented with its existing drug/compound, protein, and indication libraries. These integrated networks were reduced to a "multiscale interactomic signature" for each compound that describe its functional behavior as vectors of real values. These signatures are then used for relating compounds to each other with the hypothesis that similar signatures yield similar behavior. Our results indicated that there is significant biological information captured within our networks (particularly via side effects) which enhance the performance of our platform, as evaluated by performing all-against-all leave-one-out drug-indication association benchmarking as well as generating novel drug candidates for colon cancer and migraine disorders corroborated via literature search. Further, drug impacts on pathways derived from computed compound-protein interaction scores served as the features for a random forest machine learning model trained to predict drug-indication associations, with applications to mental disorders and cancer metastasis highlighted. This interactomic pipeline highlights the ability of Computational Analysis of Novel Drug Opportunities to accurately relate drugs in a multitarget and multiscale context, particularly for generating putative drug candidates using the information gleaned from indirect data such as side effect profiles and protein pathway information.
Collapse
Affiliation(s)
| | | | - Ram Samudrala
- Jacobs School of Medicine and Biomedical Sciences, Department of Biomedical Informatics, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
4
|
Glycogen Synthase Kinase 3: Ion Channels, Plasticity, and Diseases. Int J Mol Sci 2022; 23:ijms23084413. [PMID: 35457230 PMCID: PMC9028019 DOI: 10.3390/ijms23084413] [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] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3) is a multifaceted serine/threonine (S/T) kinase expressed in all eukaryotic cells. GSK3β is highly enriched in neurons in the central nervous system where it acts as a central hub for intracellular signaling downstream of receptors critical for neuronal function. Unlike other kinases, GSK3β is constitutively active, and its modulation mainly involves inhibition via upstream regulatory pathways rather than increased activation. Through an intricate converging signaling system, a fine-tuned balance of active and inactive GSK3β acts as a central point for the phosphorylation of numerous primed and unprimed substrates. Although the full range of molecular targets is still unknown, recent results show that voltage-gated ion channels are among the downstream targets of GSK3β. Here, we discuss the direct and indirect mechanisms by which GSK3β phosphorylates voltage-gated Na+ channels (Nav1.2 and Nav1.6) and voltage-gated K+ channels (Kv4 and Kv7) and their physiological effects on intrinsic excitability, neuronal plasticity, and behavior. We also present evidence for how unbalanced GSK3β activity can lead to maladaptive plasticity that ultimately renders neuronal circuitry more vulnerable, increasing the risk for developing neuropsychiatric disorders. In conclusion, GSK3β-dependent modulation of voltage-gated ion channels may serve as an important pharmacological target for neurotherapeutic development.
Collapse
|
5
|
El-Mallakh RS, Sampath VP, Horesh N, Lichtstein D. Endogenous Cardiac Steroids in Bipolar Disorder: State of the Art. Int J Mol Sci 2022; 23:ijms23031846. [PMID: 35163766 PMCID: PMC8836531 DOI: 10.3390/ijms23031846] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 02/04/2023] Open
Abstract
Bipolar disorder (BD) is a severe psychiatric illness with a poor prognosis and problematic, suboptimal, treatments. Treatments, borne of an understanding of the pathoetiologic mechanisms, need to be developed in order to improve outcomes. Dysregulation of cationic homeostasis is the most reproducible aspect of BD pathophysiology. Correction of ionic balance is the universal mechanism of action of all mood stabilizing medications. Endogenous sodium pump modulators (collectively known as endogenous cardiac steroids, ECS) are steroids which are synthesized in and released from the adrenal gland and brain. These compounds, by activating or inhibiting Na+, K+-ATPase activity and activating intracellular signaling cascades, have numerous effects on cell survival, vascular tone homeostasis, inflammation, and neuronal activity. For the past twenty years we have addressed the hypothesis that the Na+, K+-ATPase-ECS system may be involved in the etiology of BD. This is a focused review that presents a comprehensive model pertaining to the role of ECS in the etiology of BD. We propose that alterations in ECS metabolism in the brain cause numerous biochemical changes that underlie brain dysfunction and mood symptoms. This is based on both animal models and translational human results. There are data that demonstrate that excess ECS induce abnormal mood and activity in animals, while a specific removal of ECS with antibodies normalizes mood. There are also data indicating that circulating levels of ECS are lower in manic individuals, and that patients with BD are unable to upregulate synthesis of ECS under conditions that increase their elaboration in non-psychiatric controls. There is strong evidence for the involvement of ion dysregulation and ECS function in bipolar illness. Additional research is required to fully characterize these abnormalities and define future clinical directions.
Collapse
Affiliation(s)
- Rif S. El-Mallakh
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Correspondence: (R.S.E.-M.); (D.L.)
| | - Vishnu Priya Sampath
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research, Israel-Canada, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel; (V.P.S.); (N.H.)
| | - Noa Horesh
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research, Israel-Canada, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel; (V.P.S.); (N.H.)
| | - David Lichtstein
- Department of Medical Neurobiology, Faculty of Medicine, The Institute for Medical Research, Israel-Canada, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel; (V.P.S.); (N.H.)
- Correspondence: (R.S.E.-M.); (D.L.)
| |
Collapse
|
6
|
Clifton NE, Collado-Torres L, Burke EE, Pardiñas AF, Harwood JC, Di Florio A, Walters JTR, Owen MJ, O'Donovan MC, Weinberger DR, Holmans PA, Jaffe AE, Hall J. Developmental Profile of Psychiatric Risk Associated With Voltage-Gated Cation Channel Activity. Biol Psychiatry 2021; 90:399-408. [PMID: 33965196 PMCID: PMC8375582 DOI: 10.1016/j.biopsych.2021.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Recent breakthroughs in psychiatric genetics have implicated biological pathways onto which genetic risk for psychiatric disorders converges. However, these studies do not reveal the developmental time point(s) at which these pathways are relevant. METHODS We aimed to determine the relationship between psychiatric risk and developmental gene expression relating to discrete biological pathways. We used postmortem RNA sequencing data (BrainSeq and BrainSpan) from brain tissue at multiple prenatal and postnatal time points, with summary statistics from recent genome-wide association studies of schizophrenia, bipolar disorder, and major depressive disorder. We prioritized gene sets for overall enrichment of association with each disorder and then tested the relationship between the association of their constituent genes with their relative expression at each developmental stage. RESULTS We observed relationships between the expression of genes involved in voltage-gated cation channel activity during early midfetal, adolescence, and early adulthood time points and association with schizophrenia and bipolar disorder, such that genes more strongly associated with these disorders had relatively low expression during early midfetal development and higher expression during adolescence and early adulthood. The relationship with schizophrenia was strongest for the subset of genes related to calcium channel activity, while for bipolar disorder, the relationship was distributed between calcium and potassium channel activity genes. CONCLUSIONS Our results indicate periods during development when biological pathways related to the activity of calcium and potassium channels may be most vulnerable to the effects of genetic variants conferring risk for psychiatric disorders. Furthermore, they indicate key time points and potential targets for disorder-specific therapeutic interventions.
Collapse
Affiliation(s)
- Nicholas E Clifton
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom.
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Emily E Burke
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Janet C Harwood
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Arianna Di Florio
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael J Owen
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Departments of Psychiatry, Neurology, Neuroscience and Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Peter A Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland; Centre for Computational Biology, Johns Hopkins University Medical Campus, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
7
|
Logan RW, Ozburn AR, Arey RN, Ketchesin KD, Winquist A, Crain A, Tobe BTD, Becker-Krail D, Jarpe MB, Xue X, Zong W, Huo Z, Parekh PK, Zhu X, Fitzgerald E, Zhang H, Oliver-Smith J, DePoy LM, Hildebrand MA, Snyder EY, Tseng GC, McClung CA. Valproate reverses mania-like behaviors in mice via preferential targeting of HDAC2. Mol Psychiatry 2021; 26:4066-4084. [PMID: 33235333 PMCID: PMC8141541 DOI: 10.1038/s41380-020-00958-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/20/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Valproate (VPA) has been used in the treatment of bipolar disorder since the 1990s. However, the therapeutic targets of VPA have remained elusive. Here we employ a preclinical model to identify the therapeutic targets of VPA. We find compounds that inhibit histone deacetylase proteins (HDACs) are effective in normalizing manic-like behavior, and that class I HDACs (e.g., HDAC1 and HDAC2) are most important in this response. Using an RNAi approach, we find that HDAC2, but not HDAC1, inhibition in the ventral tegmental area (VTA) is sufficient to normalize behavior. Furthermore, HDAC2 overexpression in the VTA prevents the actions of VPA. We used RNA sequencing in both mice and human induced pluripotent stem cells (iPSCs) derived from bipolar patients to further identify important molecular targets. Together, these studies identify HDAC2 and downstream targets for the development of novel therapeutics for bipolar mania.
Collapse
Affiliation(s)
- Ryan W. Logan
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Angela R. Ozburn
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA.,VA Portland Health Care System, Portland, OR 97239, USA
| | - Rachel N. Arey
- Department of Molecular and Cellular Biology and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kyle D. Ketchesin
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Alicia Winquist
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Andrew Crain
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Brian T. D. Tobe
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.,Department of Psychiatry, Veterans Administration Medical Center, La Jolla, CA 92037, USA
| | - Darius Becker-Krail
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Matthew B. Jarpe
- Regenacy Pharmaceuticals, 303 Wyman St, Suite 300, Waltham, MA, 02451, USA
| | - Xiangning Xue
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Wei Zong
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA
| | - Puja K. Parekh
- Brain and Mind Research Institute, Department of Psychiatry, and Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xiyu Zhu
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Neuroscience, University of Pittsburgh, PA, 15260, USA
| | - Ethan Fitzgerald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Hui Zhang
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Peking Union Medical College Hospital, Beijing, China 100730
| | - Jeffrey Oliver-Smith
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Lauren M. DePoy
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Mariah A. Hildebrand
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Evan Y. Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.,Department of Pediatrics, University of California San Diego, La Jolla, CA, 92037, USA
| | - George C. Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Colleen A. McClung
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Corresponding Author: Colleen A. McClung, Ph.D., Department of Psychiatry, 450 Technology Drive, Suite 223, Pittsburgh, PA 15219, , 412-624-5547
| |
Collapse
|
8
|
El-Mallakh RS, Gao Y, You P. Role of endogenous ouabain in the etiology of bipolar disorder. Int J Bipolar Disord 2021; 9:6. [PMID: 33523310 PMCID: PMC7851255 DOI: 10.1186/s40345-020-00213-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Background Bipolar disorder is a severe psychiatric illness with poor prognosis and problematic and suboptimal treatments. Understanding the pathoetiologic mechanisms may improve treatment and outcomes. Discussion Dysregulation of cationic homeostasis is the most reproducible aspect of bipolar pathophysiology. Correction of ionic balance is the universal mechanism of action of all mood stabilizing medications. Recent discoveries of the role of endogenous sodium pump modulators (which include ‘endogenous ouabain’) in regulation of sodium and potassium distribution, inflammation, and activation of key cellular second messenger systems that are important in cell survival, and the demonstration that these stress-responsive chemicals may be dysregulated in bipolar patients, suggest that these compounds may be candidates for the coupling of environmental stressors and illness onset. Specifically, individuals with bipolar disorder appear to be unable to upregulate endogenous ouabain under conditions that require it, and therefore may experience a relative deficiency of this important regulatory hormone. In the absence of elevated endogenous ouabain, neurons are unable to maintain their normal resting potential, become relatively depolarized, and are then susceptible to inappropriate activation. Furthermore, sodium pump activity appears to be necessary to prevent inflammatory signals within the central nervous system. Nearly all available data currently support this model, but additional studies are required to solidify the role of this system. Conclusion Endogenous ouabain dysregulation appears to be a reasonable candidate for understanding the pathophysiology of bipolar disorder.
Collapse
Affiliation(s)
- Rif S El-Mallakh
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, 401 East Chestnut Street, Suite 610, Louisville, KY, 40202, USA.
| | - Yonglin Gao
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, 401 East Chestnut Street, Suite 610, Louisville, KY, 40202, USA
| | - Pan You
- Xiamen Xianyue Hospital, 399 Xianyue Road, Xiamen, China
| |
Collapse
|
9
|
Cabello-Arreola A, Ho AMC, Ozerdem A, Cuellar-Barboza AB, Kucuker MU, Heppelmann CJ, Charlesworth MC, Ceylan D, Stockmeier CA, Rajkowska G, Frye MA, Choi DS, Veldic M. Differential Dorsolateral Prefrontal Cortex Proteomic Profiles of Suicide Victims with Mood Disorders. Genes (Basel) 2020; 11:E256. [PMID: 32120974 PMCID: PMC7140872 DOI: 10.3390/genes11030256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/29/2022] Open
Abstract
Suicide is a major public health concern; nevertheless, its neurobiology remains unknown. An area of interest in suicide research is the dorsolateral prefrontal cortex (DLPFC). We aimed to identify altered proteins and potential biological pathways in the DLPFC of individuals who died by suicide employing mass spectrometry-based untargeted proteomics. Postmortem DLPFC from age-matched male suicide mood disorder cases (n = 5) and non-suicide mood disorder cases (n = 5) were compared. The proteins that differed between groups at false discovery rate (FDR) adjusted p-values (Benjamini-Hochberg-Yekutieli) <0.3 and Log2 fold change (FC) >|0.4| were considered statistically significant and were subjected to pathway analysis by Qiagen Ingenuity software. Thirty-three of the 5162 detected proteins showed significantly altered expression levels in the suicide cases and two of them after adjustment for body mass index. The top differentially expressed protein was potassium voltage-gated channel subfamily Q member 3 (KCNQ3) (Log2FC = -0.481, p = 2.10 × 10-09, FDR = 5.93 × 10-06), which also showed a trend to downregulation in Western blot (p = 0.045, Bonferroni adjusted p = 0.090). The most notably enriched pathway was the GABA receptor signaling pathway (p < 0.001). Here, we report a reduction trend of KCNQ3 levels in the DLPFC of male suicide victims with mood disorders. Further studies with a larger sample size and equal sex representation are needed.
Collapse
Affiliation(s)
| | - Ada Man-Choi Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Aysegul Ozerdem
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurosciences, Dokuz Eylul University, Health Sciences Institute, Izmir 35340, Turkey
- Department of Psychiatry, Dokuz Eylul University, School of Medicine, Izmir 35220, Turkey
| | - Alfredo B. Cuellar-Barboza
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Mehmet U. Kucuker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Deniz Ceylan
- Izmir University of Economics, Faculty of Medicine, Department of Psychiatry, Izmir 35330, Turkey
| | - Craig A. Stockmeier
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Doo-Sup Choi
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Neuroscience Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Marin Veldic
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Genomic Dissection of Bipolar Disorder and Schizophrenia, Including 28 Subphenotypes. Cell 2018; 173:1705-1715.e16. [PMID: 29906448 PMCID: PMC6432650 DOI: 10.1016/j.cell.2018.05.046] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 03/13/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023]
Abstract
Schizophrenia and bipolar disorder are two distinct diagnoses that share symptomology. Understanding the genetic factors contributing to the shared and disorder-specific symptoms will be crucial for improving diagnosis and treatment. In genetic data consisting of 53,555 cases (20,129 bipolar disorder [BD], 33,426 schizophrenia [SCZ]) and 54,065 controls, we identified 114 genome-wide significant loci implicating synaptic and neuronal pathways shared between disorders. Comparing SCZ to BD (23,585 SCZ, 15,270 BD) identified four genomic regions including one with disorder-independent causal variants and potassium ion response genes as contributing to differences in biology between the disorders. Polygenic risk score (PRS) analyses identified several significant correlations within case-only phenotypes including SCZ PRS with psychotic features and age of onset in BD. For the first time, we discover specific loci that distinguish between BD and SCZ and identify polygenic components underlying multiple symptom dimensions. These results point to the utility of genetics to inform symptomology and potential treatment.
Collapse
|
11
|
Bruce HA, Kochunov P, Paciga SA, Hyde CL, Chen X, Xie Z, Zhang B, Xi HS, O'Donnell P, Whelan C, Schubert CR, Bellon A, Ament SA, Shukla DK, Du X, Rowland LM, O'Neill H, Hong LE. Potassium channel gene associations with joint processing speed and white matter impairments in schizophrenia. GENES BRAIN AND BEHAVIOR 2017; 16:515-521. [PMID: 28188958 DOI: 10.1111/gbb.12372] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/14/2017] [Accepted: 02/07/2017] [Indexed: 12/17/2022]
Abstract
Patients with schizophrenia show decreased processing speed on neuropsychological testing and decreased white matter integrity as measured by diffusion tensor imaging, two traits shown to be both heritable and genetically associated indicating that there may be genes that influence both traits as well as schizophrenia disease risk. The potassium channel gene family is a reasonable candidate to harbor such a gene given the prominent role potassium channels play in the central nervous system in signal transduction, particularly in myelinated axons. We genotyped members of the large potassium channel gene family focusing on putatively functional single nucleotide polymorphisms (SNPs) in a population of 363 controls, 194 patients with schizophrenia spectrum disorder (SSD) and 28 patients with affective disorders with psychotic features who completed imaging and neuropsychological testing. We then performed three association analyses using three phenotypes - processing speed, whole-brain white matter fractional anisotropy (FA) and schizophrenia spectrum diagnosis. We extracted SNPs showing an association at a nominal P value of <0.05 with all three phenotypes in the expected direction: decreased processing speed, decreased FA and increased risk of SSD. A single SNP, rs8234, in the 3' untranslated region of voltage-gated potassium channel subfamily Q member 1 (KCNQ1) was identified. Rs8234 has been shown to affect KCNQ1 expression levels, and KCNQ1 levels have been shown to affect neuronal action potentials. This exploratory analysis provides preliminary data suggesting that KCNQ1 may contribute to the shared risk for diminished processing speed, diminished white mater integrity and increased risk of schizophrenia.
Collapse
Affiliation(s)
- H A Bruce
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - P Kochunov
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - S A Paciga
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - C L Hyde
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - X Chen
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - Z Xie
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - B Zhang
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - H S Xi
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - P O'Donnell
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | - C Whelan
- Pfizer Inc., Worldwide Research and Development, Cambridge, MA
| | | | - A Bellon
- Department of Psychiatry, Penn State Hershey Medical Center, Hershey, PA, USA
| | - S A Ament
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - D K Shukla
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - X Du
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - L M Rowland
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - H O'Neill
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - L E Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
12
|
Luca A, Calandra C, Luca M. Gsk3 Signalling and Redox Status in Bipolar Disorder: Evidence from Lithium Efficacy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3030547. [PMID: 27630757 PMCID: PMC5007367 DOI: 10.1155/2016/3030547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/28/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022]
Abstract
Objective. To discuss the link between glycogen synthase kinase-3 (GSK3) and the main biological alterations demonstrated in bipolar disorder (BD), with special attention to the redox status and the evidence supporting the efficacy of lithium (a GSK3 inhibitor) in the treatment of BD. Methods. A literature research on the discussed topics, using Pubmed and Google Scholar, has been conducted. Moreover, a manual selection of interesting references from the identified articles has been performed. Results. The main biological alterations of BD, pertaining to inflammation, oxidative stress, membrane ion channels, and circadian system, seem to be intertwined. The dysfunction of the GSK3 signalling pathway is involved in all the aforementioned "biological causes" of BD. In a complex scenario, it can be seen as the common denominator linking them all. Lithium inhibition of GSK3 could, at least in part, explain its positive effect on these biological dysfunctions and its superiority in terms of clinical efficacy. Conclusions. Deepening the knowledge on the molecular bases of BD is fundamental to identifying the biochemical pathways that must be targeted in order to provide patients with increasingly effective therapeutic tools against an invalidating disorder such as BD.
Collapse
Affiliation(s)
- Antonina Luca
- Department of Medical and Surgical Sciences and Advanced Technologies, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| | - Carmela Calandra
- Psychiatry Unit, Department of Medical and Surgical Sciences and Advanced Technologies, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| | - Maria Luca
- Psychiatry Unit, Department of Medical and Surgical Sciences and Advanced Technologies, University Hospital Policlinico-Vittorio Emanuele, Santa Sofia Street 78, Catania, 95100 Sicily, Italy
| |
Collapse
|
13
|
Askland KD. Editorial: "Ion channels and mental illness: exploring etiology and pathophysiology in major psychiatric disorders". Front Genet 2015; 6:152. [PMID: 25964797 PMCID: PMC4408861 DOI: 10.3389/fgene.2015.00152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/02/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Kathleen D Askland
- Psychiatry, Research and Academics, Waypoint Centre for Mental Health Care/University of Toronto Penetanguishene, ON, Canada
| |
Collapse
|
14
|
Balaraman Y, Lahiri DK, Nurnberger JI. Variants in Ion Channel Genes Link Phenotypic Features of Bipolar Illness to Specific Neurobiological Process Domains. MOLECULAR NEUROPSYCHIATRY 2015; 1:23-35. [PMID: 27602355 DOI: 10.1159/000371886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022]
Abstract
Recent advances in genome-wide association studies are pointing towards a major role for voltage-gated ion channels in neuropsychiatric disorders and, in particular, bipolar disorder (BD). The phenotype of BD is complex, with symptoms during mood episodes and deficits persisting between episodes. We have tried to elucidate the common neurobiological mechanisms associated with ion channel signaling in order to provide a new perspective on the clinical symptoms and possible endophenotypes seen in BD patients. We propose a model in which the multiple variants in genes coding for ion channel proteins would perturb motivational circuits, synaptic plasticity, myelination, hypothalamic-pituitary-adrenal axis function, circadian neuronal rhythms, and energy regulation. These changes in neurobiological mechanisms would manifest in endophenotypes of aberrant reward processing, white matter hyperintensities, deficits in executive function, altered frontolimbic connectivity, increased amygdala activity, increased melatonin suppression, decreased REM latency, and aberrant myo-inositol/ATP shuttling. The endophenotypes result in behaviors of poor impulse control, motivational changes, cognitive deficits, abnormal stress response, sleep disturbances, and energy changes involving different neurobiological process domains. The hypothesis is that these disturbances start with altered neural circuitry during development, following which multiple environmental triggers may disrupt the neuronal excitability balance through an activity-dependent molecular process, resulting in clinical mood episodes.
Collapse
Affiliation(s)
- Yokesh Balaraman
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - Debomoy K Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - John I Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| |
Collapse
|
15
|
31-year-old female shows marked improvement in depression, agitation, and panic attacks after genetic testing was used to inform treatment. Case Rep Psychiatry 2014; 2014:842349. [PMID: 24744941 PMCID: PMC3976904 DOI: 10.1155/2014/842349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 01/28/2014] [Indexed: 12/25/2022] Open
Abstract
This case describes a 31-year-old female Caucasian patient with complaints of ongoing depression, agitation, and severe panic attacks. The patient was untreated until a recent unsuccessful trial of citalopram followed by venlafaxine which produced a partial response. Genetic testing was performed to assist in treatment decisions and revealed the patient to be heterozygous for polymorphisms in 5HT2C, ANK3, and MTHFR and homozygous for a polymorphism in SLC6A4 and the low activity (Met/Met) COMT allele. In response to genetic results and clinical presentation, venlafaxine was maintained and lamotrigine was added leading to remission of agitation and depression.
Collapse
|