1
|
Mousavinejad SN, Hosseini SA, Mohammadpour M, Ferdosi F, Dadgostar E, Abdolghaderi S, Khatami SH. Long non-coding RNAs in schizophrenia. Clin Chim Acta 2025; 574:120340. [PMID: 40311728 DOI: 10.1016/j.cca.2025.120340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical regulators of the pathogenesis of schizophrenia, a complex neuropsychiatric disorder influenced by genetic and environmental factors. These transcripts modulate gene expression through diverse mechanisms, including chromatin remodeling, transcriptional regulation, and posttranscriptional modifications. Recent studies have demonstrated significant alterations in lncRNA expression profiles in both the peripheral blood and brain tissues of schizophrenia patients, highlighting their potential as biomarkers and therapeutic targets. Dysregulated lncRNAs such as Gomafu, DISC-2, BDNF-AS, MEG3, and TUG1 have been linked to neurodevelopmental processes, inflammatory responses, and key synaptic plasticity pathways implicated in schizophrenia. Furthermore, antipsychotic treatments have been shown to influence lncRNA expression, which is correlated with symptom improvement. Sex-specific and age-related differences in lncRNA regulation further underscore their complexity and relevance to schizophrenia pathophysiology. This review consolidates current knowledge on the role of lncRNAs in schizophrenia, emphasizing their diagnostic potential.
Collapse
Affiliation(s)
- Seyyed Navid Mousavinejad
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Hosseini
- Department of Neurosurgery, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mozhdeh Mohammadpour
- Department of Physical Medicine and Rehabilitation, Iran University of Medical sciences, Tehran, Iran
| | - Felora Ferdosi
- Department of Radiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Siavash Abdolghaderi
- Department of Physical Medicine and Rehabilitation, Iran University of Medical sciences, Tehran, Iran.
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Kelly ED, Ranek MJ, Zhang M, Kass DA, Muller GK. Phosphodiesterases: Evolving Concepts and Implications for Human Therapeutics. Annu Rev Pharmacol Toxicol 2025; 65:415-441. [PMID: 39322437 DOI: 10.1146/annurev-pharmtox-031524-025239] [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] [Indexed: 09/27/2024]
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides. While the 11 PDE subfamilies share common features, key differences confer signaling specificity. The differences include substrate selectivity, enzymatic activity regulation, tissue expression, and subcellular localization. Selective inhibitors of each subfamily have elucidated the protean role of PDEs in normal cell function. PDEs are also linked to diseases, some of which affect the immune, cardiac, and vascular systems. Selective PDE inhibitors are clinically used to treat these specific disorders. Ongoing preclinical studies and clinical trials are likely to lead to the approval of additional PDE-targeting drugs for therapy in human disease. In this review, we discuss the structure and function of PDEs and examine current and evolving therapeutic uses of PDE inhibitors, highlighting their mechanisms and innovative applications that could further leverage this crucial family of enzymes in clinical settings.
Collapse
Affiliation(s)
- Evan D Kelly
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA;
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manling Zhang
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Grace K Muller
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA;
| |
Collapse
|
3
|
Lv Y, Wen L, Hu WJ, Deng C, Ren HW, Bao YN, Su BW, Gao P, Man ZY, Luo YY, Li CJ, Xiang ZX, Wang B, Luan ZL. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis 2024; 39:147-171. [PMID: 37542622 DOI: 10.1007/s11011-023-01271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting 1% of the world population and ranks as one of the disorders providing the most severe burden for society. Schizophrenia etiology remains obscure involving multi-risk factors, such as genetic, environmental, nutritional, and developmental factors. Complex interactions of genetic and environmental factors have been implicated in the etiology of schizophrenia. This review provides an overview of the historical origins, pathophysiological mechanisms, diagnosis, clinical symptoms and corresponding treatment of schizophrenia. In addition, as schizophrenia is a polygenic, genetic disorder caused by the combined action of multiple micro-effective genes, we further detail several approaches, such as candidate gene association study (CGAS) and genome-wide association study (GWAS), which are commonly used in schizophrenia genomics studies. A number of GWASs about schizophrenia have been performed with the hope to identify novel, consistent and influential risk genetic factors. Finally, some schizophrenia susceptibility genes have been identified and reported in recent years and their biological functions are also listed. This review may serve as a summary of past research on schizophrenia genomics and susceptibility genes (NRG1, DISC1, RELN, BDNF, MSI2), which may point the way to future schizophrenia genetics research. In addition, depending on the above discovery of susceptibility genes and their exact function, the development and application of antipsychotic drugs will be promoted in the future.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chong Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zi-Yue Man
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, 116000, China.
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
4
|
Aftanas LI, Filimonova EA, Anisimenko MS, Berdyugina DA, Rezakova MV, Simutkin GG, Bokhan NA, Ivanova SA, Danilenko KV, Lipina TV. The habenular volume and PDE7A allelic polymorphism in major depressive disorder: preliminary findings. World J Biol Psychiatry 2023; 24:223-232. [PMID: 35673941 DOI: 10.1080/15622975.2022.2086297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES The habenula is a brain structure implicated in depression, yet with unknown molecular mechanisms. Several phosphodiesterases (PDEs) have been associated with a risk of depression. Although the role of PDE7A in the brain is unknown, it has enriched expression in the medial habenula, suggesting that it may play a role in depression. METHODS We analysed: (1) habenula volume assessed by 3-T magnetic resonance imaging (MRI) in 84 patients with major depressive disorder (MDD) and 41 healthy controls; (2) frequencies of 10 single nucleotide polymorphisms (SNPs) in PDE7A gene in 235 patients and 41 controls; and (3) both indices in 80 patients and 27 controls. The analyses considered gender, age, body mass index and season of the MRI examination. RESULTS The analysis did not reveal habenula volumetric changes in MDD patients regardless of PDE7A SNPs. However, in the combined group, the carriers of one or more mutations among 10 SNPs in the PDE7A gene had a lower volume of the left habenula (driven mainly by rs972362 and rs138599850 mutations) and consequently had the reduced habenular laterality index in comparison with individuals without PDE7A mutations. CONCLUSIONS Our findings suggest the implication of the PDE7A gene into mechanisms determining the habenula structure.
Collapse
Affiliation(s)
- Lyubomir I Aftanas
- Institute of Neurosciences and Medicine, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | | | | | | | | | - German G Simutkin
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Nikolay A Bokhan
- National Research Tomsk State University, Tomsk, Russia.,Siberian State Medical University, Tomsk, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia.,Siberian State Medical University, Tomsk, Russia
| | | | | |
Collapse
|
5
|
Targeting phosphodiesterase 4 as a therapeutic strategy for cognitive improvement. Bioorg Chem 2022; 130:106278. [DOI: 10.1016/j.bioorg.2022.106278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/22/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
|
6
|
Chen D, Wang X, Huang T, Jia J. Sleep and Late-Onset Alzheimer's Disease: Shared Genetic Risk Factors, Drug Targets, Molecular Mechanisms, and Causal Effects. Front Genet 2022; 13:794202. [PMID: 35656316 PMCID: PMC9152224 DOI: 10.3389/fgene.2022.794202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/23/2022] [Indexed: 12/30/2022] Open
Abstract
Late-onset Alzheimer's disease (AD) is associated with sleep-related phenotypes (SRPs). The fact that whether they share a common genetic etiology remains largely unknown. We explored the shared genetics and causality between AD and SRPs by using high-definition likelihood (HDL), cross-phenotype association study (CPASSOC), transcriptome-wide association study (TWAS), and bidirectional Mendelian randomization (MR) in summary-level data for AD (N = 455,258) and summary-level data for seven SRPs (sample size ranges from 359,916 to 1,331,010). AD shared a strong genetic basis with insomnia (r g = 0.20; p = 9.70 × 10-5), snoring (r g = 0.13; p = 2.45 × 10-3), and sleep duration (r g = -0.11; p = 1.18 × 10-3). The CPASSOC identifies 31 independent loci shared between AD and SRPs, including four novel shared loci. Functional analysis and the TWAS showed shared genes were enriched in liver, brain, breast, and heart tissues and highlighted the regulatory roles of immunological disorders, very-low-density lipoprotein particle clearance, triglyceride-rich lipoprotein particle clearance, chylomicron remnant clearance, and positive regulation of T-cell-mediated cytotoxicity pathways. Protein-protein interaction analysis identified three potential drug target genes (APOE, MARK4, and HLA-DRA) that interacted with known FDA-approved drug target genes. The CPASSOC and TWAS demonstrated three regions 11p11.2, 6p22.3, and 16p11.2 may account for the shared basis between AD and sleep duration or snoring. MR showed insomnia had a causal effect on AD (ORIVW = 1.02, P IVW = 6.7 × 10-6), and multivariate MR suggested a potential role of sleep duration and major depression in this association. Our findings provide strong evidence of shared genetics and causation between AD and sleep abnormalities and advance our understanding of the genetic overlap between them. Identifying shared drug targets and molecular pathways can be beneficial for treating AD and sleep disorders more efficiently.
Collapse
Affiliation(s)
- Dongze Chen
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xinpei Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China.,Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China.,Center for Statistical Science, Peking University, Beijing, China
| |
Collapse
|
7
|
Rhoades R, Solomon S, Johnson C, Teng S. Impact of SARS-CoV-2 on Host Factors Involved in Mental Disorders. Front Microbiol 2022; 13:845559. [PMID: 35444632 PMCID: PMC9014212 DOI: 10.3389/fmicb.2022.845559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/14/2022] [Indexed: 11/23/2022] Open
Abstract
COVID-19, caused by SARS-CoV-2, is a systemic illness due to its multiorgan effects in patients. The disease has a detrimental impact on respiratory and cardiovascular systems. One early symptom of infection is anosmia or lack of smell; this implicates the involvement of the olfactory bulb in COVID-19 disease and provides a route into the central nervous system. However, little is known about how SARS-CoV-2 affects neurological or psychological symptoms. SARS-CoV-2 exploits host receptors that converge on pathways that impact psychological symptoms. This systemic review discusses the ways involved by coronavirus infection and their impact on mental health disorders. We begin by briefly introducing the history of coronaviruses, followed by an overview of the essential proteins to viral entry. Then, we discuss the downstream effects of viral entry on host proteins. Finally, we review the literature on host factors that are known to play critical roles in neuropsychiatric symptoms and mental diseases and discuss how COVID-19 could impact mental health globally. Our review details the host factors and pathways involved in the cellular mechanisms, such as systemic inflammation, that play a significant role in the development of neuropsychological symptoms stemming from COVID-19 infection.
Collapse
Affiliation(s)
- Raina Rhoades
- Department of Biology, Howard University, Washington, DC, United States
| | - Sarah Solomon
- Department of Biology, Howard University, Washington, DC, United States
| | - Christina Johnson
- Department of Biology, Howard University, Washington, DC, United States
| | | |
Collapse
|
8
|
Cools R, Arnsten AFT. Neuromodulation of prefrontal cortex cognitive function in primates: the powerful roles of monoamines and acetylcholine. Neuropsychopharmacology 2022; 47:309-328. [PMID: 34312496 PMCID: PMC8617291 DOI: 10.1038/s41386-021-01100-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
The primate prefrontal cortex (PFC) subserves our highest order cognitive operations, and yet is tremendously dependent on a precise neurochemical environment for proper functioning. Depletion of noradrenaline and dopamine, or of acetylcholine from the dorsolateral PFC (dlPFC), is as devastating as removing the cortex itself, and serotonergic influences are also critical to proper functioning of the orbital and medial PFC. Most neuromodulators have a narrow inverted U dose response, which coordinates arousal state with cognitive state, and contributes to cognitive deficits with fatigue or uncontrollable stress. Studies in monkeys have revealed the molecular signaling mechanisms that govern the generation and modulation of mental representations by the dlPFC, allowing dynamic regulation of network strength, a process that requires tight regulation to prevent toxic actions, e.g., as occurs with advanced age. Brain imaging studies in humans have observed drug and genotype influences on a range of cognitive tasks and on PFC circuit functional connectivity, e.g., showing that catecholamines stabilize representations in a baseline-dependent manner. Research in monkeys has already led to new treatments for cognitive disorders in humans, encouraging future research in this important field.
Collapse
Affiliation(s)
- Roshan Cools
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
Veselinović T, Neuner I. Progress and Pitfalls in Developing Agents to Treat Neurocognitive Deficits Associated with Schizophrenia. CNS Drugs 2022; 36:819-858. [PMID: 35831706 PMCID: PMC9345797 DOI: 10.1007/s40263-022-00935-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 12/11/2022]
Abstract
Cognitive impairments associated with schizophrenia (CIAS) represent a central element of the symptomatology of this severe mental disorder. CIAS substantially determine the disease prognosis and hardly, if at all, respond to treatment with currently available antipsychotics. Remarkably, all drugs presently approved for the treatment of schizophrenia are, to varying degrees, dopamine D2/D3 receptor blockers. In turn, rapidly growing evidence suggests the immense significance of systems other than the dopaminergic system in the genesis of CIAS. Accordingly, current efforts addressing the unmet needs of patients with schizophrenia are primarily based on interventions in other non-dopaminergic systems. In this review article, we provide a brief overview of the available evidence on the importance of specific systems in the development of CIAS. In addition, we describe the promising targets for the development of new drugs that have been used so far. In doing so, we present the most important candidates that have been investigated in the field of the specific systems in recent years and present a summary of the results available at the time of drafting this review (May 2022), as well as the currently ongoing studies.
Collapse
Affiliation(s)
- Tanja Veselinović
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- JARA-BRAIN, Aachen, Germany
| |
Collapse
|
10
|
Cognitive Deficit in Schizophrenia: From Etiology to Novel Treatments. Int J Mol Sci 2021; 22:ijms22189905. [PMID: 34576069 PMCID: PMC8468549 DOI: 10.3390/ijms22189905] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 01/09/2023] Open
Abstract
Schizophrenia is a major mental illness characterized by positive and negative symptoms, and by cognitive deficit. Although cognitive impairment is disabling for patients, it has been largely neglected in the treatment of schizophrenia. There are several reasons for this lack of treatments for cognitive deficit, but the complexity of its etiology-in which neuroanatomic, biochemical and genetic factors concur-has contributed to the lack of effective treatments. In the last few years, there have been several attempts to develop novel drugs for the treatment of cognitive impairment in schizophrenia. Despite these efforts, little progress has been made. The latest findings point to the importance of developing personalized treatments for schizophrenia which enhance neuroplasticity, and of combining pharmacological treatments with non-pharmacological measures.
Collapse
|
11
|
Arnsten AFT, Datta D, Wang M. The genie in the bottle-magnified calcium signaling in dorsolateral prefrontal cortex. Mol Psychiatry 2021; 26:3684-3700. [PMID: 33319854 PMCID: PMC8203737 DOI: 10.1038/s41380-020-00973-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Neurons in the association cortices are particularly vulnerable in cognitive disorders such as schizophrenia and Alzheimer's disease, while those in primary visual cortex remain relatively resilient. This review proposes that the special molecular mechanisms needed for higher cognitive operations confer vulnerability to dysfunction, atrophy, and neurodegeneration when regulation is lost due to genetic and/or environmental insults. Accumulating data suggest that higher cortical circuits rely on magnified levels of calcium (from NMDAR, calcium channels, and/or internal release from the smooth endoplasmic reticulum) near the postsynaptic density to promote the persistent firing needed to maintain, manipulate, and store information without "bottom-up" sensory stimulation. For example, dendritic spines in the primate dorsolateral prefrontal cortex (dlPFC) express the molecular machinery for feedforward, cAMP-PKA-calcium signaling. PKA can drive internal calcium release and promote calcium flow through NMDAR and calcium channels, while in turn, calcium activates adenylyl cyclases to produce more cAMP-PKA signaling. Excessive levels of cAMP-calcium signaling can have a number of detrimental effects: for example, opening nearby K+ channels to weaken synaptic efficacy and reduce neuronal firing, and over a longer timeframe, driving calcium overload of mitochondria to induce inflammation and dendritic atrophy. Thus, calcium-cAMP signaling must be tightly regulated, e.g., by agents that catabolize cAMP or inhibit its production (PDE4, mGluR3), and by proteins that bind calcium in the cytosol (calbindin). Many genetic or inflammatory insults early in life weaken the regulation of calcium-cAMP signaling and are associated with increased risk of schizophrenia (e.g., GRM3). Age-related loss of regulatory proteins which result in elevated calcium-cAMP signaling over a long lifespan can additionally drive tau phosphorylation, amyloid pathology, and neurodegeneration, especially when protective calcium binding proteins are lost from the cytosol. Thus, the "genie" we need for our remarkable cognitive abilities may make us vulnerable to cognitive disorders when we lose essential regulation.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
12
|
Attili D, Schill DJ, DeLong CJ, Lim KC, Jiang G, Campbell KF, Walker K, Laszczyk A, McInnis MG, O'Shea KS. Astrocyte-Derived Exosomes in an iPSC Model of Bipolar Disorder. ADVANCES IN NEUROBIOLOGY 2020; 25:219-235. [PMID: 32578149 DOI: 10.1007/978-3-030-45493-7_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bipolar I Disorder (BP) is a serious, recurrent mood disorder that is characterized by alternating episodes of mania and depression. To begin to identify novel approaches and pathways involved in BP, we have obtained skin samples from BP patients and undiagnosed control (C) individuals, reprogrammed them to form induced pluripotent stem cells (iPSC), and then differentiated the stem cells into astrocytes. RNAs from BP and C astrocytes were extracted and RNAseq analysis carried out. 501 differentially expressed genes were identified, including genes for cytoskeletal elements, extracellular matrix, signaling pathways, neurodegeneration, and notably transcripts that identify exosomes. When we compared highly expressed genes using hierarchial cluster analysis, "Exosome" was the first and most highly significant cluster identified, p < 5 × 10-13, Benjamini correction. Exosomes are membrane-bound vesicles that package and remove toxic proteins from cells and also enable cell to cell communication. They carry genetic material, including DNA, mRNA and microRNAs, proteins, and lipids to target cells throughout the body. Exosomes are released by cortical neurons and astrocytes in culture and are present in BP vs C postmortem brain tissue. Little is known about what transcripts and proteins are targeted to neurons, how they regulate biological functions of the acceptor cell, or how that may be altered in mood disorders. Since astrocyte-derived exosomes have been suggested to promote neuronal plasticity, as well as to remove toxic proteins in the brain, alterations in their function or content may be involved in neurodevelopmental, neuropathological, and neuropsychiatric conditions. To examine exosome cargos and interactions with neural precursor cells, astrocytes were differentiated from four bipolar disorder (BP) and four control (C) iPSC lines. Culture supernatants from these astrocytes were collected, and exosomes isolated by ultra-centrifugation. Western blot analysis demonstrated the presence of the exosome markers CD9, CD81, and Hsp70. Nanosight technology was used to characterize exosomes from each astrocyte cell line, suggesting that exosomes were slightly more concentrated in culture supernatants derived from BP compared with C astrocytes but there was no difference in the mean sizes of the exosomes. Analysis of their function in neuronal differentiation is being carried out by labeling exosomes derived from bipolar patient and control astrocytes and adding them to control neural progenitor cells. Given the current interest in clearing toxic proteins from brains of patients with neurodegenerative disorders, exosomes may present similar opportunities in BP.
Collapse
Affiliation(s)
- D Attili
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - D J Schill
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - C J DeLong
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K C Lim
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - G Jiang
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K F Campbell
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - K Walker
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - A Laszczyk
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - M G McInnis
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - K S O'Shea
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA.
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Jankowska A, Satała G, Partyka A, Wesołowska A, Bojarski AJ, Pawłowski M, Chłoń-Rzepa G. Discovery and Development of Non-Dopaminergic Agents for the Treatment of Schizophrenia: Overview of the Preclinical and Early Clinical Studies. Curr Med Chem 2019; 26:4885-4913. [PMID: 31291870 DOI: 10.2174/0929867326666190710172002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023]
Abstract
Schizophrenia is a chronic psychiatric disorder that affects about 1 in 100 people around the world and results in persistent emotional and cognitive impairments. Untreated schizophrenia leads to deterioration in quality of life and premature death. Although the clinical efficacy of dopamine D2 receptor antagonists against positive symptoms of schizophrenia supports the dopamine hypothesis of the disease, the resistance of negative and cognitive symptoms to these drugs implicates other systems in its pathophysiology. Many studies suggest that abnormalities in glutamate homeostasis may contribute to all three groups of schizophrenia symptoms. Scientific considerations also include disorders of gamma-aminobutyric acid-ergic and serotonergic neurotransmissions as well as the role of the immune system. The purpose of this review is to update the most recent reports on the discovery and development of non-dopaminergic agents that may reduce positive, negative, and cognitive symptoms of schizophrenia, and may be alternative to currently used antipsychotics. This review collects the chemical structures of representative compounds targeting metabotropic glutamate receptor, gamma-aminobutyric acid type A receptor, alpha 7 nicotinic acetylcholine receptor, glycine transporter type 1 and glycogen synthase kinase 3 as well as results of in vitro and in vivo studies indicating their efficacy in schizophrenia. Results of clinical trials assessing the safety and efficacy of the tested compounds have also been presented. Finally, attention has been paid to multifunctional ligands with serotonin receptor affinity or phosphodiesterase inhibitory activity as novel strategies in the search for dedicated medicines for patients with schizophrenia.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Anna Partyka
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
14
|
Malakhova AV, Rudko OI, Sobolev VV, Tretiakov AV, Naumova EA, Kokaeva ZG, Azimova JE, Klimov EA. PDE4B gene polymorphism in Russian patients with panic disorder. AIMS GENETICS 2019; 6:55-63. [PMID: 31663033 PMCID: PMC6803789 DOI: 10.3934/genet.2019.3.55] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/03/2019] [Indexed: 01/16/2023]
Abstract
Background Panic disorder is a complex disease of unclear etiology but with an apparent genetic component. PDE4B gene product is involved in many cell processes owing to its function-regulation of the level of a second messenger cAMP. PDE4B gene polymorphism has been shown to be associated with some mental disorders including panic disorder. Aims The goal of our study was to evaluate the role of 3 SNPs in the PDE4B gene in the development of panic disorder. Methods 94 patients diagnosed with panic disorder according to the DSM-IV criteria were enrolled in the study. The population control group included 192 subjects. Genotyping was carried out by real-time PCR with TaqMan probes. Results The investigated substitutions are not associated with panic disorder in general and in female/male cohorts (p > 0.05). The analysis of complex genotypes demonstrated two protective complex genotypes (rs1040716:A, T + rs10454453:A + rs502958:A and rs1040716:A, T + rs502958:A) associated with panic disorder in general regardless of the patient's gender (p < 0.05). These genotypes did not correlate with the patient's sex. Conclusions We found two complex protective genotypes associated with panic disorder. This can be due to the fact that predisposition to the disease are associated with other genes, while PDE4B gene polymorphism reduces their effect.
Collapse
Affiliation(s)
- Alena V Malakhova
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Olga I Rudko
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Vladimir V Sobolev
- I.I. Mechnikov Research Institute for Vaccines and Sera, Laboratory of molecular immunology, Moscow, Russia.,University Diagnostic Laboratory LLC, Moscow, Russia.,Centre of Theoretical Problems of Physico-Chemical Pharmacology, Laboratory of Physicochemical and Genetic Problems of Dermatology, Russian Academy of Sciences, Moscow, Russia
| | | | - Elena A Naumova
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Zarema G Kokaeva
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Julia E Azimova
- University headache clinic LLC, Moscow, Russia.,The Institute of General Pathology and Pathophysiology, Laboratory of Fundamental and Applied Pain Problems, Moscow, Russia
| | - Eugene A Klimov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia.,University Diagnostic Laboratory LLC, Moscow, Russia.,Center of Experimental Embryology and Reproductive Biotechnologies, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
15
|
Lu JY, Tiwari AK, Zai GC, Rastogi A, Shaikh SA, Müller DJ, Voineskos AN, Potkin SG, Lieberman JA, Meltzer HY, Remington G, Wong AH, Kennedy JL, Zai CC. Association study of Disrupted-In-Schizophrenia-1 gene variants and tardive dyskinesia. Neurosci Lett 2018; 686:17-22. [DOI: 10.1016/j.neulet.2018.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/26/2018] [Accepted: 08/08/2018] [Indexed: 01/19/2023]
|
16
|
Alteration of Neuronal Excitability and Short-Term Synaptic Plasticity in the Prefrontal Cortex of a Mouse Model of Mental Illness. J Neurosci 2017; 37:4158-4180. [PMID: 28283561 DOI: 10.1523/jneurosci.4345-15.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 01/28/2023] Open
Abstract
Using a genetic mouse model that faithfully recapitulates a DISC1 genetic alteration strongly associated with schizophrenia and other psychiatric disorders, we examined the impact of this mutation within the prefrontal cortex. Although cortical layering, cytoarchitecture, and proteome were found to be largely unaffected, electrophysiological examination of the mPFC revealed both neuronal hyperexcitability and alterations in short-term synaptic plasticity consistent with enhanced neurotransmitter release. Increased excitability of layer II/III pyramidal neurons was accompanied by consistent reductions in voltage-activated potassium currents near the action potential threshold as well as by enhanced recruitment of inputs arising from superficial layers to layer V. We further observed reductions in both the paired-pulse ratios and the enhanced short-term depression of layer V synapses arising from superficial layers consistent with enhanced neurotransmitter release at these synapses. Recordings from layer II/III pyramidal neurons revealed action potential widening that could account for enhanced neurotransmitter release. Significantly, we found that reduced functional expression of the voltage-dependent potassium channel subunit Kv1.1 substantially contributes to both the excitability and short-term plasticity alterations that we observed. The underlying dysregulation of Kv1.1 expression was attributable to cAMP elevations in the PFC secondary to reduced phosphodiesterase 4 activity present in Disc1 deficiency and was rescued by pharmacological blockade of adenylate cyclase. Our results demonstrate a potentially devastating impact of Disc1 deficiency on neural circuit function, partly due to Kv1.1 dysregulation that leads to a dual dysfunction consisting of enhanced neuronal excitability and altered short-term synaptic plasticity.SIGNIFICANCE STATEMENT Schizophrenia is a profoundly disabling psychiatric illness with a devastating impact not only upon the afflicted but also upon their families and the broader society. Although the underlying causes of schizophrenia remain poorly understood, a growing body of studies has identified and strongly implicated various specific risk genes in schizophrenia pathogenesis. Here, using a genetic mouse model, we explored the impact of one of the most highly penetrant schizophrenia risk genes, DISC1, upon the medial prefrontal cortex, the region believed to be most prominently dysfunctional in schizophrenia. We found substantial derangements in both neuronal excitability and short-term synaptic plasticity-parameters that critically govern neural circuit information processing-suggesting that similar changes may critically, and more broadly, underlie the neural computational dysfunction prototypical of schizophrenia.
Collapse
|
17
|
Rasti B, Schaduangrat N, Shahangian SS, Nantasenamat C. Exploring the origin of phosphodiesterase inhibition via proteochemometric modeling. RSC Adv 2017. [DOI: 10.1039/c7ra02332d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A proteochemometric study of a set of phosphodiesterase 4B and 4D inhibitors sheds light on the origin of their inhibition and selectivities.
Collapse
Affiliation(s)
- Behnam Rasti
- Department of Microbiology
- Faculty of Basic Sciences
- Lahijan Branch
- Islamic Azad University (IAU)
- Lahijan
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics
- Faculty of Medical Technology
- Mahidol University
- Bangkok 10700
- Thailand
| | - S. Shirin Shahangian
- Department of Biology
- Faculty of Sciences
- University of Guilan
- Rasht 41938-33697
- Iran
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics
- Faculty of Medical Technology
- Mahidol University
- Bangkok 10700
- Thailand
| |
Collapse
|
18
|
Ferulic acid prevents LPS-induced up-regulation of PDE4B and stimulates the cAMP/CREB signaling pathway in PC12 cells. Acta Pharmacol Sin 2016; 37:1543-1554. [PMID: 27665850 DOI: 10.1038/aps.2016.88] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/01/2016] [Indexed: 12/19/2022]
Abstract
AIM Phosphodiesterase 4 (PDE4) isozymes are involved in different functions, depending on their patterns of distribution in the brain. The PDE4 subtypes are distributed in different inflammatory cells, and appear to be important regulators of inflammatory processes. In this study we examined the effects of ferulic acid (FA), a plant component with strong anti-oxidant and anti-inflammatory activities, on lipopolysaccharide (LPS)-induced up-regulation of phosphodiesterase 4B (PDE4B) in PC12 cells, which in turn regulated cellular cAMP levels and the cAMP/cAMP response element binding protein (CREB) pathway in the cells. METHODS PC12 cells were treated with LPS (1 μg/mL) for 8 h, and the changes of F-actin were detected using laser scanning confocal microscopy. The levels of pro-inflammatory cytokines were measured suing ELISA kits, and PDE4B-specific enzymatic activity was assessed with a PDE4B assay kit. The mRNA levels of PDE4B were analyzed with Q-PCR, and the protein levels of CREB and phosphorylated CREB (pCREB) were determined using immunoblotting. Furthermore, molecular docking was used to identify the interaction between PDE4B2 and FA. RESULTS Treatment of PC12 cells with LPS induced thick bundles of actin filaments appearing in the F-actin cytoskeleton, which were ameliorated by pretreatment with FA (10-40 μmol/L) or with a PDE4B inhibitor rolipram (30 μmol/L). Pretreatment with FA dose-dependently inhibited the LPS-induced production of TNF-α and IL-1β in PC12 cells. Furthermore, pretreatment with FA dose-dependently attenuated the LPS-induced up-regulation of PDE4 activity in PC12 cells. Moreover, pretreatment with FA decreased LPS-induced up-regulation of the PDE4B mRNA, and reversed LPS-induced down-regulation of CREB and pCREB in PC12 cells. The molecular docking results revealed electrostatic and hydrophobic interactions between FA and PDE4B2. CONCLUSION The beneficial effects of FA in PC12 cells might be conferred through inhibition of LPS-induced up-regulation of PDE4B and stimulation of cAMP/CREB signaling pathway. Therefore, FA may be a potential therapeutic intervention for the treatment of neuroinflammatory diseases such as AD.
Collapse
|
19
|
Phillips WA, Larkum ME, Harley CW, Silverstein SM. The effects of arousal on apical amplification and conscious state. Neurosci Conscious 2016; 2016:niw015. [PMID: 29877512 PMCID: PMC5934888 DOI: 10.1093/nc/niw015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/28/2016] [Accepted: 08/08/2016] [Indexed: 01/19/2023] Open
Abstract
Neocortical pyramidal cells can integrate two classes of input separately and use one to modulate response to the other. Their tuft dendrites are electrotonically separated from basal dendrites and soma by the apical dendrite, and apical hyperpolarization-activated currents (Ih) further isolate subthreshold integration of tuft inputs. When apical depolarization exceeds a threshold, however, it can enhance response to the basal inputs that specify the cell's selective sensitivity. This process is referred to as apical amplification (AA). We review evidence suggesting that, by regulating Ih in the apical compartments, adrenergic arousal controls the coupling between apical and somatic integration zones thus modifying cognitive capabilities closely associated with consciousness. Evidence relating AA to schizophrenia, sleep, and anesthesia is reviewed, and we assess theories that emphasize the relevance of AA to consciousness. Implications for theories of neocortical computation that emphasize context-sensitive modulation are summarized. We conclude that the findings concerning AA and its regulation by arousal offer a new perspective on states of consciousness, the function and evolution of neocortex, and psychopathology. Many issues worthy of closer examination arise.
Collapse
Affiliation(s)
- W. A. Phillips
- School of Natural Sciences, University of Stirling, Scotland FK9 4LA, UK
| | - M. E. Larkum
- Neurocure Cluster of Excellence, Department of Biology, Humboldt University,
Charitéplatz 1, Berlin 10117, Germany
| | - C. W. Harley
- Psychology Department, Memorial University of Newfoundland, St. John's, NL A1C 5S7,
P.O. Box 4200, Canada
| | | |
Collapse
|
20
|
Borkowska M, Millar JK, Price DJ. Altered Disrupted-in-Schizophrenia-1 Function Affects the Development of Cortical Parvalbumin Interneurons by an Indirect Mechanism. PLoS One 2016; 11:e0156082. [PMID: 27244370 PMCID: PMC4886955 DOI: 10.1371/journal.pone.0156082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/09/2016] [Indexed: 01/01/2023] Open
Abstract
Disrupted-in-Schizophrenia-1 (DISC1) gene has been linked to schizophrenia and related major mental illness. Mouse Disc1 has been implicated in brain development, mainly in the proliferation, differentiation, lamination, neurite outgrowth and synapse formation and maintenance of cortical excitatory neurons. Here, the effects of two loss-of-function point mutations in the mouse Disc1 sequence (Q31L and L100P) on cortical inhibitory interneurons were investigated. None of the mutations affected the overall number of interneurons. However, the 100P, but not the 31L, mutation resulted in a significant decrease in the numbers of interneurons expressing parvalbumin mRNA and protein across the sensory cortex. To investigate role of Disc1 in regulation of parvalbumin expression, mouse wild-type Disc-1 or the 100P mutant form were electroporated in utero into cortical excitatory neurons. Overexpression of wild-type Disc1 in these cells caused increased densities of parvalbumin-expressing interneurons in the electroporated area and in areas connected with it, whereas expression of Disc1-100P did not. We conclude that the 100P mutation prevents expression of parvalbumin by a normally sized cohort of interneurons and that altering Disc1 function in cortical excitatory neurons indirectly affects parvalbumin expression by cortical interneurons, perhaps as a result of altered functional input from the excitatory neurons.
Collapse
Affiliation(s)
- Malgorzata Borkowska
- University of Edinburgh Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh EH8 9XD, United Kingdom
- * E-mail:
| | - J. Kirsty Millar
- University of Edinburgh Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, Crewe Road, Edinburgh EH4 2XU, United Kingdom
| | - David J. Price
- University of Edinburgh Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
21
|
Phosphodiesterase4D (PDE4D)--A risk factor for atrial fibrillation and stroke? J Neurol Sci 2015; 359:266-74. [PMID: 26671126 DOI: 10.1016/j.jns.2015.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/23/2023]
Abstract
Mutations in the gene encoding phosphodiesterase 4D (PDE4D) enzyme are associated with ischemic stroke; however the functional implications of such mutations are not well understood. PDE4D is part of a complex protein family modulating intracellular signalling by cyclic nucleotides. The PDE4 family includes subtypes A-D, all of which show unique intracellular, cellular and tissue distribution. PDE4D is the major subtype expressed in human atrial myocytes and involved in the pathophysiology of arrhythmias, such as atrial fibrillation. The PDE4D enzyme hydrolyses cyclic adenosine monophosphate (cAMP). Though diverging results are reported, several population based studies describe association of various PDE4D single nucleotide polymorphisms (SNP) with cardio-embolic stroke in particular. Functionally, a down regulation of PDE4D variants has been reported in stroke patients. The anti-inflammatory and vasodilator properties of PDE4 inhibitors make them suitable for treatment of stroke and cardiovascular disease. PDE4D has recently been suggested as factor in atrial fibrillation. This review summarizes the possible function of PDE4D in the brain, heart, and vasculature. Further, association of the described SNPs, in particular, with cardioembolic stroke, is reviewed. Current findings on the PDE4D mutations suggest functionality involves an increased cardiac risk factor as well as augmented risk of atrial fibrillation.
Collapse
|
22
|
Lundegaard PR, Anastasaki C, Grant NJ, Sillito RR, Zich J, Zeng Z, Paranthaman K, Larsen AP, Armstrong JD, Porteous DJ, Patton EE. MEK Inhibitors Reverse cAMP-Mediated Anxiety in Zebrafish. ACTA ACUST UNITED AC 2015; 22:1335-46. [PMID: 26388333 PMCID: PMC4623357 DOI: 10.1016/j.chembiol.2015.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 08/11/2015] [Accepted: 08/14/2015] [Indexed: 12/14/2022]
Abstract
Altered phosphodiesterase (PDE)-cyclic AMP (cAMP) activity is frequently associated with anxiety disorders, but current therapies act by reducing neuronal excitability rather than targeting PDE-cAMP-mediated signaling pathways. Here, we report the novel repositioning of anti-cancer MEK inhibitors as anxiolytics in a zebrafish model of anxiety-like behaviors. PDE inhibitors or activators of adenylate cyclase cause behaviors consistent with anxiety in larvae and adult zebrafish. Small-molecule screening identifies MEK inhibitors as potent suppressors of cAMP anxiety behaviors in both larvae and adult zebrafish, while causing no anxiolytic behavioral effects on their own. The mechanism underlying cAMP-induced anxiety is via crosstalk to activation of the RAS-MAPK signaling pathway. We propose that targeting crosstalk signaling pathways can be an effective strategy for mental health disorders, and advance the repositioning of MEK inhibitors as behavior stabilizers in the context of increased cAMP.
Collapse
Affiliation(s)
- Pia R Lundegaard
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Department of Biomedical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Corina Anastasaki
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Nicola J Grant
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Rowland R Sillito
- Actual Analytics Ltd, 2.05 Wilkie Building, 22-23 Teviot Row, Edinburgh EH8 9AG, UK
| | - Judith Zich
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Zhiqiang Zeng
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karthika Paranthaman
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Anders Peter Larsen
- Department of Biomedical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, 2200 Copenhagen, Denmark
| | - J Douglas Armstrong
- Actual Analytics Ltd, 2.05 Wilkie Building, 22-23 Teviot Row, Edinburgh EH8 9AG, UK; School of Informatics, Institute for Adaptive and Neural Computation, Informatics Forum, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | - E Elizabeth Patton
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.
| |
Collapse
|
23
|
Zhang X, Li X, Li M, Ren J, Yun K, An Y, Lin L, Zhang H. Venlafaxine increases cell proliferation and regulates DISC1, PDE4B and NMDA receptor 2B expression in the hippocampus in chronic mild stress mice. Eur J Pharmacol 2015; 755:58-65. [PMID: 25769842 DOI: 10.1016/j.ejphar.2015.02.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/19/2015] [Accepted: 02/24/2015] [Indexed: 10/23/2022]
Abstract
Recent evidence has identified disrupted in schizophrenia-1 (DISC1) as an important genetic risk factor for the development of many psychiatric disorders, including major depressive disorders. In addition, studies using animal models have demonstrated that chronic stress affects hippocampal structure and function. However, the functional effects of chronic stress on DISC1 remain unknown. Using a chronic mild stress (CMS) paradigm, we investigated the effects of CMS on depressive-like behaviors, hippocampal cell proliferation, and hippocampal protein expression of DISC1, phosphodiesterase 4B (PDE4B) and N-methyl-d-aspartate receptor 2B subunit (NMDA receptor 2B), which may be involved in the regulation of DISC1 and neurogenesis. We also examined the effects and possible mechanisms of the antidepressant venlafaxine in CMS mice. CMS increased the expression of DISC1 and PDE4B. Chronic treatment with venlafaxine blocked the increases in these proteins, and also reversed the CMS-induced decrease in neurogenesis and NMDA receptor 2B protein in the hippocampus. These results suggest that DISC1 may play an important role in the etiology of depression and in the action of antidepressants.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Min Li
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jintao Ren
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ke Yun
- Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yan An
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| | - Lei Lin
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| | - Hailong Zhang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
24
|
Abstract
Glia are starting to be accepted as the equal of neurons. Their impact on intelligence, environmental enrichment, and cerebral dominance forms the basis for understanding the role of glia in stress. Along with neurons, astrocytes, microglia, NG2 cells, and oligodendrocytes all contribute. Glia can even be protective against drug abuse. Glial effects on depression, mood disorders and schizophrenia are reviewed.
Collapse
|
25
|
El-Hassar L, Simen AA, Duque A, Patel KD, Kaczmarek LK, Arnsten AF, Yeckel MF. Disrupted in schizophrenia 1 modulates medial prefrontal cortex pyramidal neuron activity through cAMP regulation of transient receptor potential C and small-conductance K+ channels. Biol Psychiatry 2014; 76:476-85. [PMID: 24560582 PMCID: PMC4104266 DOI: 10.1016/j.biopsych.2013.12.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 12/22/2013] [Accepted: 12/30/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Disrupted in schizophrenia 1 (DISC1) is a protein implicated in schizophrenia, bipolar disorder, major depressive disorder, and autism. To date, most of research examining DISC1 function has focused on its role in neurodevelopment, despite its presence throughout life. DISC1 also regulates cyclic adenosine monophosphate (cAMP) signaling by increasing type 4 phosphodiesterase catabolism of cAMP when cAMP concentrations are high. In this study, we tested the hypothesis that DISC1, through its regulation of cAMP, modulates I-SK and I-TRPC channel-mediated ionic currents that we have shown previously to regulate the activity of mature prefrontal cortical pyramidal neurons. METHODS We used patch-clamp recordings in prefrontal cortical slices from adult rats in which DISC1 function was reduced in vivo by short hairpin RNA viral knockdown or in vitro by dialysis of DISC1 antibodies. RESULTS We found that DISC1 disruption resulted in an increase of metabotropic glutamate receptor-induced intracellular calcium (Ca2+) waves, small-conductance K+ (SK)-mediated hyperpolarization and a decrease of transient receptor potential C (TRPC)-mediated sustained depolarization. Consistent with a role for DISC1 in regulation of cAMP signaling, forskolin-induced cAMP production also increased intracellular Ca2+ waves, I-SK and decreased I-TRPC. Lastly, inhibiting cAMP generation with guanfacine, an α2A-noradrenergic agonist, normalized the function of SK and TRPC channels. CONCLUSIONS Based on our findings, we propose that diminished DISC1 function, such as occurs in some mental disorders, can lead to the disruption of normal patterns of prefrontal cortex activity through the loss of cAMP regulation of metabotropic glutamate receptor-mediated intracellular Ca2+ waves, SK and TRPC channel activity.
Collapse
Affiliation(s)
- Lynda El-Hassar
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut.
| | - Arthur A. Simen
- Department of Psychiatry, Yale University School of Medicine
| | - Alvaro Duque
- Department of Neurobiology, Yale University School of Medicine
| | - Kiran D. Patel
- Department of Psychiatry, Yale University School of Medicine
| | | | - Amy F.T. Arnsten
- Department of Neurobiology, Yale University School of Medicine,The Kavli Institute for Neuroscience, Yale University School of Medicine
| | - Mark F. Yeckel
- Department of Neurobiology, Yale University School of Medicine,The Kavli Institute for Neuroscience, Yale University School of Medicine
| |
Collapse
|
26
|
Ahmad F, Murata T, Shimizu K, Degerman E, Maurice D, Manganiello V. Cyclic nucleotide phosphodiesterases: important signaling modulators and therapeutic targets. Oral Dis 2014; 21:e25-50. [PMID: 25056711 DOI: 10.1111/odi.12275] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 02/06/2023]
Abstract
By catalyzing hydrolysis of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), cyclic nucleotide phosphodiesterases are critical regulators of their intracellular concentrations and their biological effects. As these intracellular second messengers control many cellular homeostatic processes, dysregulation of their signals and signaling pathways initiate or modulate pathophysiological pathways related to various disease states, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication, chronic obstructive pulmonary disease, and psoriasis. Alterations in expression of PDEs and PDE-gene mutations (especially mutations in PDE6, PDE8B, PDE11A, and PDE4) have been implicated in various diseases and cancer pathologies. PDEs also play important role in formation and function of multimolecular signaling/regulatory complexes, called signalosomes. At specific intracellular locations, individual PDEs, together with pathway-specific signaling molecules, regulators, and effectors, are incorporated into specific signalosomes, where they facilitate and regulate compartmentalization of cyclic nucleotide signaling pathways and specific cellular functions. Currently, only a limited number of PDE inhibitors (PDE3, PDE4, PDE5 inhibitors) are used in clinical practice. Future paths to novel drug discovery include the crystal structure-based design approach, which has resulted in generation of more effective family-selective inhibitors, as well as burgeoning development of strategies to alter compartmentalized cyclic nucleotide signaling pathways by selectively targeting individual PDEs and their signalosome partners.
Collapse
Affiliation(s)
- F Ahmad
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
27
|
Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 2014; 13:290-314. [PMID: 24687066 DOI: 10.1038/nrd4228] [Citation(s) in RCA: 593] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) catalyse the hydrolysis of cyclic AMP and cyclic GMP, thereby regulating the intracellular concentrations of these cyclic nucleotides, their signalling pathways and, consequently, myriad biological responses in health and disease. Currently, a small number of PDE inhibitors are used clinically for treating the pathophysiological dysregulation of cyclic nucleotide signalling in several disorders, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication and chronic obstructive pulmonary disease. However, pharmaceutical interest in PDEs has been reignited by the increasing understanding of the roles of individual PDEs in regulating the subcellular compartmentalization of specific cyclic nucleotide signalling pathways, by the structure-based design of novel specific inhibitors and by the development of more sophisticated strategies to target individual PDE variants.
Collapse
Affiliation(s)
- Donald H Maurice
- Biomedical and Molecular Sciences, Queen's University, Kingston K7L3N6, Ontario, Canada
| | - Hengming Ke
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Faiyaz Ahmad
- Cardiovascular and Pulmonary Branch, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Yousheng Wang
- Beijing Technology and Business University, Beijing 100048, China
| | - Jay Chung
- Genetics and Developmental Biology Center, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
28
|
Otero C, Peñaloza JP, Rodas PI, Fernández-Ramires R, Velasquez L, Jung JE. Temporal and spatial regulation of cAMP signaling in disease: role of cyclic nucleotide phosphodiesterases. Fundam Clin Pharmacol 2014; 28:593-607. [PMID: 24750474 DOI: 10.1111/fcp.12080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/28/2014] [Accepted: 04/17/2014] [Indexed: 01/19/2023]
Abstract
Since its discovery, cAMP has been proposed as one of the most versatile second messengers. The remarkable feature of cAMP to tightly control highly diverse physiological processes, including metabolism, homeostasis, secretion, muscle contraction, cell proliferation and migration, immune response, and gene transcription, is reflected by millions of different articles worldwide. Compartmentalization of cAMP in space and time, maintained by mainly phosphodiesterases, contributes to the maintenance of equilibrium inside the cell where one signal can trigger many different events. Novel cAMP sensors seem to carry out certain unexpected signaling properties of cAMP and thereby to permit delicate adaptations of biologic responses. Measuring space and time events with biosensors will increase our current knowledge on the pathophysiology of diseases, such as chronic obstructive pulmonary disease, asthma, cognitive impairment, cancer, and renal and heart failure. Further insights into the cAMP dynamics will help to optimize the pharmacological treatment for these diseases.
Collapse
Affiliation(s)
- Carolina Otero
- Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile; Centro para el Desarrollo de la Nanociencia y Nanotecnologia, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
29
|
Sheppard CL, Lee LCY, Hill EV, Henderson DJP, Anthony DF, Houslay DM, Yalla KC, Cairns LS, Dunlop AJ, Baillie GS, Huston E, Houslay MD. Mitotic activation of the DISC1-inducible cyclic AMP phosphodiesterase-4D9 (PDE4D9), through multi-site phosphorylation, influences cell cycle progression. Cell Signal 2014; 26:1958-74. [PMID: 24815749 DOI: 10.1016/j.cellsig.2014.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 10/25/2022]
Abstract
In Rat-1 cells, the dramatic decrease in the levels of both intracellular cyclic 3'5' adenosine monophosphate (cyclic AMP; cAMP) and in the activity of cAMP-activated protein kinase A (PKA) observed in mitosis was paralleled by a profound increase in cAMP hydrolyzing phosphodiesterase-4 (PDE4) activity. The decrease in PKA activity, which occurs during mitosis, was attributable to PDE4 activation as the PDE4 selective inhibitor, rolipram, but not the phosphodiesterase-3 (PDE3) inhibitor, cilostamide, specifically ablated this cell cycle-dependent effect. PDE4 inhibition caused Rat-1 cells to move from S phase into G2/M more rapidly, to transit through G2/M more quickly and to remain in G1 for a longer period. Inhibition of PDE3 elicited no observable effects on cell cycle dynamics. Selective immunopurification of each of the four PDE4 sub-families identified PDE4D as being selectively activated in mitosis. Subsequent analysis uncovered PDE4D9, an isoform whose expression can be regulated by Disrupted-In-Schizophrenia 1 (DISC1)/activating transcription factor 4 (ATF4) complex, as the sole PDE4 species activated during mitosis in Rat-1 cells. PDE4D9 becomes activated in mitosis through dual phosphorylation at Ser585 and Ser245, involving the combined action of ERK and an unidentified 'switch' kinase that has previously been shown to be activated by H2O2. Additionally, in mitosis, PDE4D9 also becomes phosphorylated at Ser67 and Ser81, through the action of MK2 (MAPKAPK2) and AMP kinase (AMPK), respectively. The multisite phosphorylation of PDE4D9 by all four of these protein kinases leads to decreased mobility (band-shift) of PDE4D9 on SDS-PAGE. PDE4D9 is predominantly concentrated in the perinuclear region of Rat-1 cells but with a fraction distributed asymmetrically at the cell margins. Our investigations demonstrate that the diminished levels of cAMP and PKA activity that characterise mitosis are due to enhanced cAMP degradation by PDE4D9. PDE4D9, was found to locate primarily not only in the perinuclear region of Rat-1 cells but also at the cell margins. We propose that the sequestration of PDE4D9 in a specific complex together with AMPK, ERK, MK2 and the H2O2-activatable 'switch' kinase allows for its selective multi-site phosphorylation, activation and regulation in mitosis.
Collapse
Affiliation(s)
- Catherine L Sheppard
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Louisa C Y Lee
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Elaine V Hill
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - David J P Henderson
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Diana F Anthony
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Daniel M Houslay
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Krishna C Yalla
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Lynne S Cairns
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Allan J Dunlop
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Elaine Huston
- Institute of Pharmaceutical Science, King's College London, 5th Floor, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Miles D Houslay
- Institute of Pharmaceutical Science, King's College London, 5th Floor, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
30
|
Regulation of N-methyl-D-aspartate receptors by disrupted-in-schizophrenia-1. Biol Psychiatry 2014; 75:414-424. [PMID: 23906531 PMCID: PMC3864617 DOI: 10.1016/j.biopsych.2013.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/12/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Genetic studies have implicated disrupted-in-schizophrenia-1 (DISC1) as a risk factor for a wide range of mental conditions, including schizophrenia. Because N-methyl-D-aspartate receptor (NMDAR) dysfunction has been strongly linked to the pathophysiology of these conditions, we examined whether the NMDAR is a potential target of DISC1. METHODS DISC1 was knocked down with a small inference RNA. NMDAR-mediated currents were recorded and NMDAR expression was measured. RESULTS We found that cellular knockdown of DISC1 significantly increased NMDAR currents in cortical cultures, which were accompanied by an increase in the expression of NMDAR subunit, GluN2A. NMDAR-mediated synaptic response in prefrontal cortical pyramidal neurons was also increased by DISC1 knockdown in vivo. The effect of DISC1 knockdown on NMDAR currents in cortical cultures was blocked by protein kinase A (PKA) inhibitor, occluded by PKA activator, and prevented by phosphodiesterase 4 inhibitor. Knockdown of DISC1 caused a significant increase of cyclic adenosine monophosphate response element-binding protein (CREB) activity. Inhibiting CREB prevented the DISC1 deficiency-induced increase of NMDAR currents and GluN2A clusters. CONCLUSIONS Our results suggest that DISC1 exerts an important impact on NMDAR expression and function through a phosphodiesterase 4/PKA/CREB-dependent mechanism, which provides a potential molecular basis for the role of DISC1 in influencing NMDAR-dependent cognitive and emotional processes.
Collapse
|
31
|
Gamo NJ, Duque A, Paspalas CD, Kata A, Fine R, Boven L, Bryan C, Lo T, Anighoro K, Bermudez L, Peng K, Annor A, Raja A, Mansson E, Taylor SR, Patel K, Simen AA, Arnsten AFT. Role of disrupted in schizophrenia 1 (DISC1) in stress-induced prefrontal cognitive dysfunction. Transl Psychiatry 2013; 3:e328. [PMID: 24301646 PMCID: PMC4030323 DOI: 10.1038/tp.2013.104] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/05/2013] [Accepted: 10/09/2013] [Indexed: 12/22/2022] Open
Abstract
Recent genetic studies have linked mental illness to alterations in disrupted in schizophrenia 1 (DISC1), a multifunctional scaffolding protein that regulates cyclic adenosine monophosphate (cAMP) signaling via interactions with phosphodiesterase 4 (PDE4). High levels of cAMP during stress exposure impair function of the prefrontal cortex (PFC), a region gravely afflicted in mental illness. As stress can aggravate mental illness, genetic insults to DISC1 may worsen symptoms by increasing cAMP levels. The current study examined whether viral knockdown (KD) of the Disc1 gene in rat PFC increases susceptibility to stress-induced PFC dysfunction. Rats were trained in a spatial working memory task before receiving infusions of (a) an active viral construct that knocked down Disc1 in PFC (DISC1 KD group), (b) a 'scrambled' construct that had no effect on Disc1 (Scrambled group), or (c) an active construct that reduced DISC1 expression dorsal to PFC (Anatomical Control group). Data were compared with an unoperated Control group. Cognitive performance was assessed following mild restraint stress that had no effect on normal animals. DISC1 KD rats were impaired by 1 h restraint stress, whereas Scrambled, Control, and Anatomical Control groups were unaffected. Thus, knocking down Disc1 in PFC reduced the threshold for stress-induced cognitive dysfunction, possibly through disinhibited cAMP signaling at neuronal network synapses. These findings may explain why patients with DISC1 mutations may be especially vulnerable to the effects of stress.
Collapse
Affiliation(s)
- N J Gamo
- Department of Neurobiology, Yale University, New Haven, CT, USA,Department of Neurobiology, Yale University, 600 N. Wolfe Street, Baltimore, MD 21287, USA. E-mail:
| | - A Duque
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - C D Paspalas
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - A Kata
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - R Fine
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - L Boven
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - C Bryan
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - T Lo
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - K Anighoro
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - L Bermudez
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - K Peng
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - A Annor
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - A Raja
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - E Mansson
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - S R Taylor
- Department of Neurobiology, Yale University, New Haven, CT, USA
| | - K Patel
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - A A Simen
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - A F T Arnsten
- Department of Neurobiology, Yale University, New Haven, CT, USA
| |
Collapse
|
32
|
Soda T, Frank C, Ishizuka K, Baccarella A, Park YU, Flood Z, Park SK, Sawa A, Tsai LH. DISC1-ATF4 transcriptional repression complex: dual regulation of the cAMP-PDE4 cascade by DISC1. Mol Psychiatry 2013; 18:898-908. [PMID: 23587879 PMCID: PMC3730299 DOI: 10.1038/mp.2013.38] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 01/12/2013] [Accepted: 01/31/2013] [Indexed: 02/08/2023]
Abstract
Disrupted-In-Schizophrenia 1 (DISC1), a risk factor for major mental illnesses, has been studied extensively in the context of neurodevelopment. However, the role of DISC1 in neuronal signaling, particularly in conjunction with intracellular cascades that occur in response to dopamine, a neurotransmitter implicated in numerous psychiatric disorders, remains elusive. Previous data suggest that DISC1 interacts with numerous proteins that impact neuronal function, including activating transcription factor 4 (ATF4). In this study, we identify a novel DISC1 and ATF4 binding region in the genomic locus of phosphodiesterase 4D (PDE4D), a gene implicated in psychiatric disorders. We found that the loss of function of either DISC1 or ATF4 increases PDE4D9 transcription, and that the association of DISC1 with the PDE4D9 locus requires ATF4. We also show that PDE4D9 is increased by D1-type dopamine receptor dopaminergic stimulation. We demonstrate that the mechanism for this increase is due to DISC1 dissociation from the PDE4D locus in mouse brain. We further characterize the interaction of DISC1 with ATF4 to show that it is regulated via protein kinase A-mediated phosphorylation of DISC1 serine-58. Our results suggest that the release of DISC1-mediated transcriptional repression of PDE4D9 acts as feedback inhibition to regulate dopaminergic signaling. Furthermore, as DISC1 loss-of-function leads to a specific increase in PDE4D9, PDE4D9 itself may represent an attractive target for therapeutic approaches in psychiatric disorders.
Collapse
Affiliation(s)
- T Soda
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA,Howard Hughes Medical Institute, Cambridge, MA, USA,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA,Daniel Tosteson Medical Education Center, Boston, MA, USA
| | - C Frank
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA,Howard Hughes Medical Institute, Cambridge, MA, USA,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - K Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Baccarella
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Y-U Park
- Division of Molecular and Life Science, Department of Life Science, Biotechnology Research Center, Pohang University of Science and Technology, Pohang, Korea
| | - Z Flood
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA,Howard Hughes Medical Institute, Cambridge, MA, USA,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - S K Park
- Division of Molecular and Life Science, Department of Life Science, Biotechnology Research Center, Pohang University of Science and Technology, Pohang, Korea
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - L-H Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA,Howard Hughes Medical Institute, Cambridge, MA, USA,Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA,Howard Hughes Medical Institute, 77 Massachusetts Avenue, Room 46-4235, Cambridge, MA 02139, USA. E-mail:
| |
Collapse
|
33
|
Hoffmann M. The human frontal lobes and frontal network systems: an evolutionary, clinical, and treatment perspective. ISRN NEUROLOGY 2013; 2013:892459. [PMID: 23577266 PMCID: PMC3612492 DOI: 10.1155/2013/892459] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/19/2012] [Indexed: 12/27/2022]
Abstract
Frontal lobe syndromes, better termed as frontal network systems, are relatively unique in that they may manifest from almost any brain region, due to their widespread connectivity. The understandings of the manifold expressions seen clinically are helped by considering evolutionary origins, the contribution of the state-dependent ascending monoaminergic neurotransmitter systems, and cerebral connectivity. Hence, the so-called networktopathies may be a better term for the syndromes encountered clinically. An increasing array of metric tests are becoming available that complement that long standing history of qualitative bedside assessments pioneered by Alexander Luria, for example. An understanding of the vast panoply of frontal systems' syndromes has been pivotal in understanding and diagnosing the most common dementia syndrome under the age of 60, for example, frontotemporal lobe degeneration. New treatment options are also progressively becoming available, with recent evidence of dopaminergic augmentation, for example, being helpful in traumatic brain injury. The latter include not only psychopharmacological options but also device-based therapies including mirror visual feedback therapy.
Collapse
Affiliation(s)
- Michael Hoffmann
- Director Stroke and Cognitive Neurology Programs, James A. Haley Veterans' Hospital, 13000 Bruce B. Down's Boulevard, Tampa, FL 33612, USA
- Cognitive Neurologist and Director SciBrain, Roskamp Neurosciences Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA
| |
Collapse
|
34
|
Hida H, Mouri A, Noda Y. Behavioral phenotypes in schizophrenic animal models with multiple combinations of genetic and environmental factors. J Pharmacol Sci 2013; 121:185-91. [PMID: 23449491 DOI: 10.1254/jphs.12r15cp] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Schizophrenia is a multifactorial psychiatric disorder in which both genetic and environmental factors play a role. Genetic [e.g., Disrupted-in-schizophrenia 1 (DISC1), Neuregulin-1 (NRG1)] and environmental factors (e.g., maternal viral infection, obstetric complications, social stress) may act during the developmental period to increase the incidence of schizophrenia. In animal models, interactions between susceptibility genes and the environment can be controlled in ways not possible in humans; therefore, such models are useful for investigating interactions between or within factors in the pathogenesis and pathophysiology of schizophrenia. We provide an overview of schizophrenic animal models investigating interactions between or within factors. First, we reviewed gene-environment interaction animal models, in which schizophrenic candidate gene mutant mice were subjected to perinatal immune activation or adolescent stress. Next, environment-environment interaction animal models, in which mice were subjected to a combination of perinatal immune activation and adolescent administration of drugs, were described. These animal models showed interaction between or within factors; behavioral changes, which were obscured by each factor, were marked by interaction of factors and vice versa. Appropriate behavioral approaches with such models will be invaluable for translational research on novel compounds, and also for providing insight into the pathogenesis and pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Hirotake Hida
- Division of Clinical Sciences and Neuropsychopharmacology, Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | | | | |
Collapse
|
35
|
Cacabelos R, Cacabelos P, Aliev G. Genomics of schizophrenia and pharmacogenomics of antipsychotic drugs. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojpsych.2013.31008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Ayalew M, Le-Niculescu H, Levey DF, Jain N, Changala B, Patel SD, Winiger E, Breier A, Shekhar A, Amdur R, Koller D, Nurnberger JI, Corvin A, Geyer M, Tsuang MT, Salomon D, Schork NJ, Fanous AH, O'Donovan MC, Niculescu AB. Convergent functional genomics of schizophrenia: from comprehensive understanding to genetic risk prediction. Mol Psychiatry 2012; 17:887-905. [PMID: 22584867 PMCID: PMC3427857 DOI: 10.1038/mp.2012.37] [Citation(s) in RCA: 309] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/28/2012] [Accepted: 03/05/2012] [Indexed: 02/07/2023]
Abstract
We have used a translational convergent functional genomics (CFG) approach to identify and prioritize genes involved in schizophrenia, by gene-level integration of genome-wide association study data with other genetic and gene expression studies in humans and animal models. Using this polyevidence scoring and pathway analyses, we identify top genes (DISC1, TCF4, MBP, MOBP, NCAM1, NRCAM, NDUFV2, RAB18, as well as ADCYAP1, BDNF, CNR1, COMT, DRD2, DTNBP1, GAD1, GRIA1, GRIN2B, HTR2A, NRG1, RELN, SNAP-25, TNIK), brain development, myelination, cell adhesion, glutamate receptor signaling, G-protein-coupled receptor signaling and cAMP-mediated signaling as key to pathophysiology and as targets for therapeutic intervention. Overall, the data are consistent with a model of disrupted connectivity in schizophrenia, resulting from the effects of neurodevelopmental environmental stress on a background of genetic vulnerability. In addition, we show how the top candidate genes identified by CFG can be used to generate a genetic risk prediction score (GRPS) to aid schizophrenia diagnostics, with predictive ability in independent cohorts. The GRPS also differentiates classic age of onset schizophrenia from early onset and late-onset disease. We also show, in three independent cohorts, two European American and one African American, increasing overlap, reproducibility and consistency of findings from single-nucleotide polymorphisms to genes, then genes prioritized by CFG, and ultimately at the level of biological pathways and mechanisms. Finally, we compared our top candidate genes for schizophrenia from this analysis with top candidate genes for bipolar disorder and anxiety disorders from previous CFG analyses conducted by us, as well as findings from the fields of autism and Alzheimer. Overall, our work maps the genomic and biological landscape for schizophrenia, providing leads towards a better understanding of illness, diagnostics and therapeutics. It also reveals the significant genetic overlap with other major psychiatric disorder domains, suggesting the need for improved nosology.
Collapse
Affiliation(s)
- M Ayalew
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N Jain
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - B Changala
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Winiger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Breier
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Amdur
- Washington DC VA Medical Center, Washington, DC, USA
| | - D Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J I Nurnberger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Corvin
- Department of Psychiatry, Trinity College, Dublin, Ireland
| | - M Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - M T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - D Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - N J Schork
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A H Fanous
- Washington DC VA Medical Center, Washington, DC, USA
| | - M C O'Donovan
- Department of Psychological Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
37
|
Wiescholleck V, Manahan-Vaughan D. PDE4 inhibition enhances hippocampal synaptic plasticity in vivo and rescues MK801-induced impairment of long-term potentiation and object recognition memory in an animal model of psychosis. Transl Psychiatry 2012; 2:e89. [PMID: 22832854 PMCID: PMC3309535 DOI: 10.1038/tp.2012.17] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inhibition of phosphodiesterase type 4 (PDE4) by rolipram (4-(3-(cyclopentyloxy)-4-methoxyphenyl)-pyrrolidin-2-one) has been the focus of many behavioral and molecular studies in the recent years. Rolipram exhibits memory-enhancing effects in rodents. In vitro studies have shown that long-term potentiation (LTP), which may comprise a cellular substrate for learning, is also enhanced by rolipram. However, effects have not been assessed in vivo. Rolipram has antipsychotic properties. Psychosis affects cognition and in animal models of psychosis LTP is impaired. In this study, we investigated if PDE4 inhibition improves LTP in healthy animals in vivo and if PDE4 inhibition rescues impaired LTP and prevents object recognition memory deficits in an animal model of psychosis. Recordings were made from the hippocampus of adult, freely behaving Wistar rats. Thirty minutes after treatment with rolipram or vehicle, a tetanus was applied to the medial perforant path to elicit short-term potentiation (STP) in the dentate gyrus. At this time-point, radioimmunoassay revealed that rolipram significantly elevated cyclic adenosine monophosphate levels in the dorsal hippocampus, in line with reports by others that rolipram mediates decreased PDE4 activity. In healthy animals, both intracerebroventricular and subcutaneous treatment with rolipram facilitated STP into LTP, suggesting that PDE4 inhibition may have a permissive role in plasticity mechanisms that are relevant for learning and memory. One week after a single systemic treatment with the irreversible N-methyl-D-aspartate antagonist, MK801, LTP and object recognition memory were significantly impaired, but could be rescued by PDE4 inhibition. These data suggest that the relief of cognitive disturbances in psychosis models by rolipram may be mediated in part by a rescue of hippocampal LTP.
Collapse
Affiliation(s)
- V Wiescholleck
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum, Germany,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - D Manahan-Vaughan
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum, Germany,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany,Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, MA 4/149, Universitaetsstr. 150, 44780 Bochum, Germany. E-mail:
| |
Collapse
|
38
|
Arnsten AFT, Jin LE. Guanfacine for the treatment of cognitive disorders: a century of discoveries at Yale. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2012; 85:45-58. [PMID: 22461743 PMCID: PMC3313539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The prefrontal cortex (PFC) is among the most evolved brain regions, contributing to our highest order cognitive abilities. It regulates behavior, thought, and emotion using working memory. Many cognitive disorders involve impairments of the PFC. A century of discoveries at Yale Medical School has revealed the neurobiology of PFC cognitive functions, as well as the molecular needs of these circuits. This work has led to the identification of therapeutic targets to treat cognitive disorders. Recent research has found that the noradrenergic α2A agonist guanfacine can improve PFC function by strengthening PFC network connections via inhibition of cAMP-potassium channel signaling in postsynaptic spines. Guanfacine is now being used to treat a variety of PFC cognitive disorders, including Tourette's Syndrome and Attention Deficit Hyperactivity Disorder (ADHD). This article reviews the history of Yale discoveries on the neurobiology of PFC working memory function and the identification of guanfacine for treating cognitive disorders.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neurobiology, Yale School of Medicine, New Haven, CT 06510, USA.
| | | |
Collapse
|
39
|
Phosphodiesterase 4 inhibition enhances the dopamine D1 receptor/PKA/DARPP-32 signaling cascade in frontal cortex. Psychopharmacology (Berl) 2012; 219:1065-79. [PMID: 21833500 PMCID: PMC3539205 DOI: 10.1007/s00213-011-2436-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 08/01/2011] [Indexed: 10/17/2022]
Abstract
RATIONALE Alteration of dopamine neurotransmission in the prefrontal cortex, especially hypofunction of dopamine D1 receptors, contributes to psychotic symptoms and cognitive deficit in schizophrenia. D1 receptors signal through the cAMP/PKA second messenger cascade, which is modulated by phosphodiesterase (PDE) enzymes that hydrolyze and inactivate cyclic nucleotides. Though several PDEs are expressed in cortical neurons, the PDE4 enzyme family (PDE4A-D) has been implicated in the control of cognitive function. The best studied isoform, PDE4B, interacts with a schizophrenia susceptibility factor, disrupted in schizophrenia 1 (DISC1). OBJECTIVES We explore the control of mouse frontal cortex dopamine D1 receptor signaling and associated behavior by PDE4. RESULTS Inhibition of PDE4 by rolipram induced activation of cAMP/PKA signaling in cortical slices and in vivo, leading to the phosphorylation of DARPP-32 and other postsynaptic and presynaptic PKA-substrates. Rolipram also enhanced DARPP-32 phosphorylation invoked by D1 receptor activation. Immunohistochemical studies demonstrated PDE4A, PDE4B, and PDE4D expression in DARPP-32-positive neurons in layer VI of frontal cortex, most likely in D1 receptor-positive, glutamatergic corticothalamic pyramidal neurons. Furthermore, the ability of rolipram treatment to improve the performance of mice in a sensorimotor gating test was DARPP-32-dependent. CONCLUSIONS PDE4, which is co-expressed with DARPP-32 in D1 receptor-positive cortical pyramidal neurons in layer VI, modulates the level of D1 receptor signaling and DARPP-32 phosphorylation in the frontal cortex, likely influencing cognitive function. These biochemical and behavioral actions of PDE4 inhibitors may contribute to the hypothesized antipsychotic actions of this class of compounds.
Collapse
|
40
|
Abstract
Schizophrenia (SCZ) is among the most disabling of mental disorders. Several neurobiological hypotheses have been postulated as responsible for SCZ pathogenesis: polygenic/multifactorial genomic defects, intrauterine and perinatal environment-genome interactions, neurodevelopmental defects, dopaminergic, cholinergic, serotonergic, gamma-aminobutiric acid (GABAergic), neuropeptidergic and glutamatergic/N-Methyl-D-Aspartate (NMDA) dysfunctions, seasonal infection, neuroimmune dysfunction, and epigenetic dysregulation. SCZ has a heritability estimated at 60-90%. Genetic studies in SCZ have revealed the presence of chromosome anomalies, copy number variants, multiple single-nucleotide polymorphisms of susceptibility distributed across the human genome, aberrant single nucleotide polymorphisms (SNPs) in microRNA genes, mitochondrial DNA mutations, and epigenetic phenomena. Pharmacogenetic studies of psychotropic drug response have focused on determining the relationship between variation in specific candidate genes and the positive and adverse effects of drug treatment. Approximately, 18% of neuroleptics are major substrates of CYP1A2 enzymes, 40% of CYP2D6, and 23% of CYP3A4; 24% of antidepressants are major substrates of CYP1A2 enzymes, 5% of CYP2B6, 38% of CYP2C19, 85% of CYP2D6, and 38% of CYP3A4; 7% of benzodiazepines are major substrates of CYP2C19 enzymes, 20% of CYP2D6, and 95% of CYP3A4. About 10-20% of Western populations are defective in genes of the CYP superfamily. Only 26% of Southern Europeans are pure extensive metabolizers for the trigenic cluster integrated by the CYP2D6+CYP2C19+CYP2C9 genes. The pharmacogenomic response of SCZ patients to conventional psychotropic drugs also depends on genetic variants associated with SCZ-related genes. Consequently, the incorporation of pharmacogenomic procedures both to drugs in development and drugs on the market would help to optimize therapeutics in SCZ and other central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, 15165-Bergondo, Coruña, Spain.
| | | |
Collapse
|
41
|
Gamo NJ, Arnsten AFT. Molecular modulation of prefrontal cortex: rational development of treatments for psychiatric disorders. Behav Neurosci 2011; 125:282-96. [PMID: 21480691 DOI: 10.1037/a0023165] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Dysfunction of the prefrontal cortex (PFC) is a central feature of many psychiatric disorders, such as attention deficit hyperactivity disorder (ADHD), posttraumatic stress disorder (PTSD), schizophrenia, and bipolar disorder. Thus, understanding molecular influences on PFC function through basic research in animals is essential to rational drug development. In this review, we discuss the molecular signaling events initiated by norepinephrine and dopamine that strengthen working memory function mediated by the dorsolateral PFC under optimal conditions, and weaken working memory function during uncontrollable stress. We also discuss how these intracellular mechanisms can be compromised in psychiatric disorders, and how novel treatments based on these findings may restore a molecular environment conducive to PFC regulation of behavior, thought and emotion. Examples of successful translation from animals to humans include guanfacine for the treatment of ADHD and related PFC disorders, and prazosin for the treatment of PTSD.
Collapse
Affiliation(s)
- Nao J Gamo
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06520-8001, USA.
| | | |
Collapse
|
42
|
Page CP, Spina D. Phosphodiesterase inhibitors in the treatment of inflammatory diseases. Handb Exp Pharmacol 2011:391-414. [PMID: 21695650 DOI: 10.1007/978-3-642-17969-3_17] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Phosphodiesterase 4 (PDE4) belongs to a family of enzymes which catalyzes the breakdown of 3, 5'-adenosine cyclic monophosphate (cAMP) and is ubiquitously expressed in inflammatory cells. There is little evidence that inflammatory diseases are caused by increased expression of this isoenzyme, although human inflammatory cell activity can be suppressed by selective PDE4 inhibitors. Consequently, there is intense interest in the development of selective PDE4 inhibitors for the treatment of a range of inflammatory diseases, including asthma, chronic obstructive pulmonary disease (COPD), inflammatory bowel disease, and psoriasis. Recent clinical trials with roflumilast in COPD have confirmed the therapeutic potential of targeting PDE4 and recently roflumilast has been approved for marketing in Europe and the USA, although side effects such as gastrointestinal disturbances, particularly nausea and emesis as well as headache and weight loss, may limit the use of this drug class, at least when administered by the oral route. However, a number of strategies are currently being pursued in attempts to improve clinical efficacy and reduce side effects of PDE4 inhibitors, including delivery via the inhaled route, development of nonemetic PDE4 inhibitors, mixed PDE inhibitors, and/or antisense biologicals targeted toward PDE4.
Collapse
Affiliation(s)
- C P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, Franklin Wilkins Building, London SE1 9NH, UK.
| | | |
Collapse
|
43
|
De Rienzo G, Bishop JA, Mao Y, Pan L, Ma TP, Moens CB, Tsai LH, Sive H. Disc1 regulates both β-catenin-mediated and noncanonical Wnt signaling during vertebrate embryogenesis. FASEB J 2011; 25:4184-97. [PMID: 21859895 DOI: 10.1096/fj.11-186239] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Disc1 is a schizophrenia risk gene that engages multiple signaling pathways during neurogenesis and brain development. Using the zebrafish as a tool, we analyze the function of zebrafish Disc1 (zDisc1) at the earliest stages of brain and body development. We define a "tool" as a biological system that gives insight into mechanisms underlying a human disorder, although the system does not phenocopy the disorder. A zDisc1 peptide binds to GSK3β, and zDisc1 directs early brain development and neurogenesis, by promoting β-catenin-mediated Wnt signaling and inhibiting GSK3β activity. zDisc1 loss-of-function embryos additionally display a convergence and extension phenotype, demonstrated by abnormal movement of dorsolateral cells during gastrulation, through changes in gene expression, and later through formation of abnormal, U-shaped muscle segments, and a truncated tail. These phenotypes are caused by alterations in the noncanonical Wnt pathway, via Daam and Rho signaling. The convergence and extension phenotype can be rescued by a dominant negative GSK3β construct, suggesting that zDisc1 inhibits GSK3β activity during noncanonical Wnt signaling. This is the first demonstration that Disc1 modulates the noncanonical Wnt pathway and suggests a previously unconsidered mechanism by which Disc1 may contribute to the etiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gianluca De Rienzo
- Whitehead institute for Biomedical Research, 9 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Verhoeven WM, Tuinier S, van der Burgt I. Top-down or bottom-up: Contrasting perspectives on psychiatric diagnoses. Biologics 2011; 2:409-17. [PMID: 19707372 PMCID: PMC2721407 DOI: 10.2147/btt.s3053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Clinical psychiatry is confronted with the expanding knowledge of medical genetics. Most of the research into the genetic underpinnings of major mental disorders as described in the categorical taxonomies, however, did reveal linkage with a variety of chromosomes. This heterogeneity of results is most probably due to the assumption that the nosological categories as used in these studies are disease entities with clear boundaries. If the reverse way of looking, the so-called bottom-up approach, is applied, it becomes clear that genetic abnormalities are in most cases not associated with a single psychiatric disorder but with a certain probability to develop a variety of aspecific psychiatric symptoms. The adequacy of the categorical taxonomy, the so-called top-down approach, seems to be inversely related to the amount of empirical etiological data. This is illustrated by four rather prevalent genetic syndromes, fragile X syndrome, Prader-Willi syndrome, 22q11 deletion syndrome, and Noonan syndrome, as well as by some cases with rare chromosomal abnormalities. From these examples, it becomes clear that psychotic symptoms as well as mood, anxiety, and autistic features can be found in a great variety of different genetic syndromes. A psychiatric phenotype exists, but comprises, apart from the chance to present several psychiatric symptoms, all elements from developmental, neurocognitive, and physical characteristics.
Collapse
|
45
|
Arnsten AFT. Prefrontal cortical network connections: key site of vulnerability in stress and schizophrenia. Int J Dev Neurosci 2011; 29:215-23. [PMID: 21345366 PMCID: PMC3115784 DOI: 10.1016/j.ijdevneu.2011.02.006] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 01/01/2023] Open
Abstract
The symptoms of schizophrenia involve profound dysfunction of the prefrontal cortex (PFC). PFC networks create our "mental sketch pad", and PFC dysfunction contributes to symptoms such as cognitive deficits, thought disorder, delusions and hallucinations. Neuropathological studies of schizophrenia have shown marked loss of dendritic spines in deep layer III, the sublayer where PFC microcircuits reside. The microcircuits consist of recurrent excitatory pyramidal cell networks that interconnect on spines, and excite each other via NMDA receptor signaling. The pyramidal cell persistent firing is sculpted by lateral inhibition from GABAergic basket and chandelier cells, thus creating tuned, persistent firing needed for accurate representational knowledge (i.e., working memory). The strength of pyramidal cell network connections is markedly and flexibly altered by intracellular signaling pathways in dendritic spines, a process called dynamic network connectivity (DNC). DNC proteins such as HCN channels are concentrated on dendritic spines in deep layer III. Under optimal conditions, network inputs to pyramidal cells are strengthened by noradrenergic alpha-2A inhibition of cAMP-HCN channel signaling, and sculpted by dopamine D1-cAMP-HCN channel weakening of inappropriate inputs. However, with stress exposure, high levels of cAMP-HCN channel signaling produces a collapse in network firing. With chronic stress exposure, spines reduce in size and are lost, and this process involves increased PKC signaling. Importantly, molecules that normally strengthen PFC networks connections and/or reverse the stress response, are often genetically altered in schizophrenia. As exposure to stress is a key factor in the precipitation of schizophrenic symptoms, these dysregulated signaling pathways in deep layer III may interact with already vulnerable circuitry to cause spine loss and the descent into illness.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neurobiology, Yale Medical School, 333 Cedar St., New Haven, CT 06510, USA.
| |
Collapse
|
46
|
Petti M, Samanich J, Pan Q, Huang CK, Reinmund J, Farooqi S, Morrow B, Babcock M. Molecular characterization of an interstitial deletion of 1p31.3 in a patient with obesity and psychiatric illness and a review of the literature. Am J Med Genet A 2011; 155A:825-32. [PMID: 21416589 DOI: 10.1002/ajmg.a.33869] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 11/22/2010] [Indexed: 01/14/2023]
Abstract
We report on the clinical and array-based characterization of an interstitial 1p31.3 deletion in a 15-year-old male patient with obesity, behavioral problems including multiple psychiatric diagnoses, mild intellectual impairment, facial dysmorphism, and a strong family history of psychiatric illness. The deletion breakpoints were determined by molecular karyotyping, revealing a 3.2 Mb excision. Patients previously reported with hemizygous deletions including this cytogenetic band had intellectual impairment and some facial features that overlap with our patient's phenotype. However, their deletions were larger, encompassing several cytogenetic bands, making this case the smallest deletion to date that we are aware of sharing these phenotypic characteristics. There are 17 genes that map to the interval. Two genes within the interval, LEPR and PDE4B, are interesting candidates for these phenotypes because of their potential role in obesity and psychiatric illness, respectively. Identification of the smaller deletion underscores the importance of combining clinical investigation and array comparative genomic hybridization analysis for appropriate diagnosis, genetic counseling and potentially for prenatal diagnosis.
Collapse
Affiliation(s)
- Marilena Petti
- Division of Translational Genetics, Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Horesh Y, Katsel P, Haroutunian V, Domany E. Gene expression signature is shared by patients with Alzheimer’s disease and schizophrenia at the superior temporal gyrus. Eur J Neurol 2011; 18:410-24. [DOI: 10.1111/j.1468-1331.2010.03166.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Phosphodiesterases in the central nervous system: implications in mood and cognitive disorders. Handb Exp Pharmacol 2011:447-85. [PMID: 21695652 DOI: 10.1007/978-3-642-17969-3_19] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a superfamily of enzymes that are involved in the regulation of the intracellular second messengers cyclic AMP (cAMP) and cyclic GMP (cGMP) by controlling their rates of hydrolysis. There are 11 different PDE families and each family typically has multiple isoforms and splice variants. The PDEs differ in their structures, distribution, modes of regulation, and sensitivity to inhibitors. Since PDEs have been shown to play distinct roles in processes of emotion and related learning and memory processes, selective PDE inhibitors, by preventing the breakdown of cAMP and/or cGMP, modulate mood and related cognitive activity. This review discusses the current state and future development in the burgeoning field of PDEs in the central nervous system. It is becoming increasingly clear that PDE inhibitors have therapeutic potential for the treatment of neuropsychiatric disorders involving disturbances of mood, emotion, and cognition.
Collapse
|
49
|
O'Connell G, Lawrie SM, McIntosh AM, Hall J. Schizophrenia risk genes: Implications for future drug development and discovery. Biochem Pharmacol 2010; 81:1367-73. [PMID: 21093417 DOI: 10.1016/j.bcp.2010.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 11/07/2010] [Accepted: 11/08/2010] [Indexed: 02/04/2023]
Abstract
Present-day development of improved treatments for schizophrenia is hindered by uncertain models of disease, inter-individual response variability in clinical trials and a paucity of sensitive measures of treatment effects. Findings from genetic research emphasize the potential for schizophrenia risk genes to help develop focused treatments, discover new drug targets and provide markers of clinical subtypes. Advances in genetic technologies also provide novel modes of drug discovery in schizophrenia such as transcriptomics, epigenetics and transgenic animal models. In this review, we discuss proven and proposed ways risk genes can be used to enhance the development and discovery of treatments for schizophrenia and highlight key studies in these approaches.
Collapse
Affiliation(s)
- Garret O'Connell
- Division of Psychiatry, University of Edinburgh, Scotland, United Kingdom.
| | | | | | | |
Collapse
|
50
|
The human area postrema and other nuclei related to the emetic reflex express cAMP phosphodiesterases 4B and 4D. J Chem Neuroanat 2010; 40:36-42. [DOI: 10.1016/j.jchemneu.2010.03.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 03/17/2010] [Accepted: 03/17/2010] [Indexed: 01/01/2023]
|