1
|
Ventura S, Mathieson SR, O'Toole JM, Livingstone V, Murray DM, Boylan GB. Infant sleep EEG features at 4 months as biomarkers of neurodevelopment at 18 months. Pediatr Res 2025:10.1038/s41390-025-03893-6. [PMID: 39979586 DOI: 10.1038/s41390-025-03893-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/10/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Sleep parameters evolve in parallel with neurodevelopment. Sleep participates in synaptic homeostasis and memory consolidation and infant sleep parameters correlate with later aspects of early childhood cognition. METHODS Typically developing, term-born infants had a diurnal sleep-EEG at 4 months and Griffiths III developmental assessment at 18 months. EEG analysis included sleep macrostructure (i.e. durations of total sleep and sleep stages, and latencies to sleep and REM), sleep spindle features, and quantitative EEG features (qEEG): interhemispheric connectivity and spectral power. We assessed the correlations between these EEG features and Griffiths III quotients. RESULTS Sleep recordings from 92 infants were analyzed. Sleep latency was positively associated with the Griffiths III Foundations of Learning subscale and N3 sleep duration was positively correlated with the Personal-Social-Emotional subscale. Sleep spindle synchrony was negatively associated with Eye and Hand Coordination, Personal-Social-Emotional, Gross Motor, and General Development quotients. Sleep spindle duration was negatively associated with the Personal-Social-Emotional and Gross Motor subscales. In some sleep states, delta 1 and 2 EEG spectral power and interhemispheric coherence measures were correlated with subscale quotients. CONCLUSION Certain sleep features in the EEG of 4-month-old infants are associated with neurodevelopment at 18 months and may be useful early biomarkers of neurodevelopment. IMPACT This study shows that the EEG during infant sleep may provide insights into later neurodevelopmental outcomes. We have examined novel EEG sleep spindle features and shown that spindle duration and synchrony may help predict neurodevelopmental outcomes. Sleep macrostructure elements such as latency to sleep, N3 duration, and qEEG features such as interhemispheric coherence and spectral power measures at 4 months may be useful for the assessment of future neurodevelopmental outcomes. Due to exceptional neuroplasticity in infancy, EEG biomarkers of neurodevelopment may support early and targeted intervention to optimize outcomes.
Collapse
Affiliation(s)
- Soraia Ventura
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics & Child Health, University College Cork, Cork, Ireland
| | - Sean R Mathieson
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics & Child Health, University College Cork, Cork, Ireland
| | - John M O'Toole
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Vicki Livingstone
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics & Child Health, University College Cork, Cork, Ireland
| | - Deirdre M Murray
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics & Child Health, University College Cork, Cork, Ireland
| | - Geraldine B Boylan
- INFANT Research Centre, University College Cork, Cork, Ireland.
- Department of Paediatrics & Child Health, University College Cork, Cork, Ireland.
| |
Collapse
|
2
|
You Y, Liu H, Yang Z, Chen Y, Yang F, Yu T, Zhang Y. Anesthetic spindles serve as EEG markers of the depth variations in anesthesia induced by multifarious general anesthetics in mouse experiments. Front Pharmacol 2024; 15:1474923. [PMID: 39734402 PMCID: PMC11671261 DOI: 10.3389/fphar.2024.1474923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Background Mice play a crucial role in studying the mechanisms of general anesthesia. However, identifying reliable EEG markers for different depths of anesthesia induced by multifarious agents remains a significant challenge. Spindle activity, typically observed during NREM sleep, reflects synchronized thalamocortical activity and is characterized by a frequency range of 7-15 Hz and a duration of 0.5-3 s. Similar patterns, referred to as "anesthetic spindles," are also observed in the EEG during general anesthesia. However, the variability of anesthetic spindles across different anesthetic agents and depths is not yet fully understood. Method Mice were anesthetized with dexmedetomidine, propofol, ketamine, etomidate, isoflurane, or sevoflurane, and cortical EEG recordings were obtained. EEG signals were bandpass filtered between 0.1 and 60 Hz and analyzed using a custom MATLAB script for spindle detection. Anesthesia depth was assessed based on Guedel's modified stages of anesthesia and the presence of burst suppression in the EEG. Results Compared to sleep spindles, anesthetic spindles induced by the different agents exhibited higher amplitudes and longer durations. Isoflurane- and sevoflurane-induced spindles varied with the depth of anesthesia. Spindles associated with etomidate were prominent during induction and light anesthesia, whereas those induced by sevoflurane and isoflurane were more dominant during deep anesthesia and emergence. Post-anesthesia, spindles persisted but ceased more quickly following inhalational anesthesia. Conclusion Anesthesia spindle waves reflect distinct changes in anesthesia depth and persist following emergence, serving as objective EEG markers for assessing both anesthesia depth and the recovery process.
Collapse
Affiliation(s)
- Ying You
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Hui Liu
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Zhanfei Yang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yuxuan Chen
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Fei Yang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Tian Yu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yu Zhang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
3
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
4
|
Ferrarelli F. Sleep spindles as neurophysiological biomarkers of schizophrenia. Eur J Neurosci 2024; 59:1907-1917. [PMID: 37885306 DOI: 10.1111/ejn.16178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/17/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder characterized by a wide range of clinical symptoms, including disrupted sleep. In recent years, there has been growing interest in assessing alterations in sleep parameters in patients with SCZ. Sleep spindles are brief (0.5-2 s) bursts of 12- to 16-Hz rhythmic electroencephalogram (EEG) oscillatory activity occurring during non-rapid eye movement (NREM) sleep. Spindles have been implicated in several critical brain functions, including learning, memory and plasticity, and are thought to reflect the integrity of underlying thalamocortical circuits. This review aims to provide an overview of the current research investigating sleep spindles in SCZ. After briefly describing the neurophysiological features of sleep spindles, I will discuss alterations in spindle characteristics observed in SCZ, their associations with the clinical symptomatology of these patients and their putative underlying neuronal and molecular mechanisms. I will then discuss the utility of sleep spindle measures as predictors of treatment response and disease progression. Finally, I will highlight future directions for research in this emerging field, including the prospect of utilizing sleep spindles as neurophysiological biomarkers of SCZ.
Collapse
Affiliation(s)
- Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Luo Y, Yu Y, He H, Fan N. Acute ketamine induces neuronal hyperexcitability and deficits in prepulse inhibition by upregulating IL-6. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110913. [PMID: 38103855 DOI: 10.1016/j.pnpbp.2023.110913] [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: 09/11/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Acute ketamine administration results in psychotic symptoms similar to those observed in schizophrenia and is regarded as a pharmacological model of schizophrenia. Accumulating evidence suggests that patients with schizophrenia show increased IL-6 levels in the blood and cerebrospinal fluid and that IL-6 levels are associated with the severity of psychotic symptoms. In the present study, we found that a single ketamine exposure led to increased expression of IL-6 and IL-6Rα, decreased dendritic spine density, increased expression and currents of T-type calcium channels, and increased neuron excitability in the hippocampal CA1 area 12 h after exposure. Acute ketamine administration also led to impaired prepulse inhibition (PPI) 12 h after administration. Additionally, we found that the expression of signaling molecules IKKα/β, NF-κB, JAK2, and STAT3 was upregulated 12 h after a single ketamine injection. The decreases in dendritic spine density, the increases in calcium currents and neuron excitability, and the impairments in PPI were ameliorated by blocking IL-6 or IL-6Rα. Our findings show that blocking IL-6 or its receptor may protect hippocampal neurons from hyperexcitability, thereby ameliorating ketamine-induced psychotic effects. Our study provides additional evidence that targeting IL-6 and its receptor is a potential strategy for treating psychotic symptoms in acute ketamine-induced psychosis and schizophrenia.
Collapse
Affiliation(s)
- Yayan Luo
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Yang Yu
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Hongbo He
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Ni Fan
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China.
| |
Collapse
|
6
|
Weiss N, Zamponi GW. The T-type calcium channelosome. Pflugers Arch 2024; 476:163-177. [PMID: 38036777 DOI: 10.1007/s00424-023-02891-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
T-type calcium channels perform crucial physiological roles across a wide spectrum of tissues, spanning both neuronal and non-neuronal system. For instance, they serve as pivotal regulators of neuronal excitability, contribute to cardiac pacemaking, and mediate the secretion of hormones. These functions significantly hinge upon the intricate interplay of T-type channels with interacting proteins that modulate their expression and function at the plasma membrane. In this review, we offer a panoramic exploration of the current knowledge surrounding these T-type channel interactors, and spotlight certain aspects of their potential for drug-based therapeutic intervention.
Collapse
Affiliation(s)
- Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
7
|
Mayeli A, Donati FL, Ferrarelli F. Altered Sleep Oscillations as Neurophysiological Biomarkers of Schizophrenia. ADVANCES IN NEUROBIOLOGY 2024; 40:351-383. [PMID: 39562451 DOI: 10.1007/978-3-031-69491-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Sleep spindles and slow waves are the two main oscillatory activities occurring during nonrapid eye movement (NREM) sleep. Here, we will first describe the electrophysiological characteristics of these sleep oscillations along with the neurophysiological and molecular mechanisms underlying their generation and synchronization in the healthy brain. We will then review the extant evidence of deficits in sleep spindles and, to a lesser extent, slow waves, including in slow wave-spindle coupling, in patients with Schizophrenia (SCZ) across the course of the disorder, from at-risk to chronic stages. Next, we will discuss how these sleep oscillatory deficits point to defects in neuronal circuits within the thalamocortical network as well as to alterations in molecular neurotransmission implicating the GABAergic and glutamatergic systems in SCZ. Finally, after explaining how spindle and slow waves may represent neurophysiological biomarkers with predictive, diagnostic, and prognostic potential, we will present novel pharmacological and neuromodulatory interventions aimed at restoring sleep oscillatory deficits in SCZ, which in turn may serve as target engagement biomarkers to ameliorate the clinical symptoms and the quality of life of individuals affected by this devastating brain disorder.
Collapse
Affiliation(s)
- Ahmad Mayeli
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
McLean RT, Buist E, St Clair D, Wei J. An examination of plasma autoantibodies against voltage gated calcium channels in schizophrenia. Brain Behav Immun Health 2023; 28:100603. [PMID: 36865984 PMCID: PMC9972490 DOI: 10.1016/j.bbih.2023.100603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Autoantibodies targeting the central nervous system have been shown to induce psychiatric symptoms resembling schizophrenia. Concurrently, genetic studies have characterised a number of risk variants associated with schizophrenia although their functional implications are largely unknown. Any biological effects of functional variants on protein function may potentially be replicated by the presence of autoantibodies against such proteins. Recent research has demonstrated that the R1346H variant in the CACNA1I gene coding for the Cav 3.3 protein results in a synaptic reduction of Cav3.3 voltage gated calcium channels and, consequently, sleep spindles, which have been shown to correlate with several symptom domains in patients with schizophrenia. The present study measured plasma levels of IgG against two peptides derived from CACNA1I and CACNA1C, respectively, in patients with schizophrenia and healthy controls. The results demonstrated that increased anti-CACNA1I IgG levels were associated with schizophrenia but not associated with any symptom domain related to the reduction of sleep spindles. In contrast to previously published work indicating that inflammation may be a marker for a depressive phenotype, plasma levels of IgG against either CACNA1I or CACNA1C peptides were not associated with depressive symptoms, suggesting that anti-Cav3.3 autoantibodies may function independently of pro-inflammatory processes.
Collapse
Affiliation(s)
- Ryan Thomas McLean
- Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
| | - Elizabeth Buist
- Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
- New Craigs Hospital, Inverness, UK
| | - David St Clair
- Department of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Jun Wei
- Institute of Health Research and Innovation, University of the Highlands and Islands, Inverness, UK
- Corresponding author. Institute of Health Research and Innovation, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK.
| |
Collapse
|
9
|
Matthews LG, Puryear CB, Correia SS, Srinivasan S, Belfort GM, Pan MK, Kuo SH. T-type calcium channels as therapeutic targets in essential tremor and Parkinson's disease. Ann Clin Transl Neurol 2023; 10:462-483. [PMID: 36738196 PMCID: PMC10109288 DOI: 10.1002/acn3.51735] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 02/05/2023] Open
Abstract
Neuronal action potential firing patterns are key components of healthy brain function. Importantly, restoring dysregulated neuronal firing patterns has the potential to be a promising strategy in the development of novel therapeutics for disorders of the central nervous system. Here, we review the pathophysiology of essential tremor and Parkinson's disease, the two most common movement disorders, with a focus on mechanisms underlying the genesis of abnormal firing patterns in the implicated neural circuits. Aberrant burst firing of neurons in the cerebello-thalamo-cortical and basal ganglia-thalamo-cortical circuits contribute to the clinical symptoms of essential tremor and Parkinson's disease, respectively, and T-type calcium channels play a key role in regulating this activity in both the disorders. Accordingly, modulating T-type calcium channel activity has received attention as a potentially promising therapeutic approach to normalize abnormal burst firing in these diseases. In this review, we explore the evidence supporting the theory that T-type calcium channel blockers can ameliorate the pathophysiologic mechanisms underlying essential tremor and Parkinson's disease, furthering the case for clinical investigation of these compounds. We conclude with key considerations for future investigational efforts, providing a critical framework for the development of much needed agents capable of targeting the dysfunctional circuitry underlying movement disorders such as essential tremor, Parkinson's disease, and beyond.
Collapse
Affiliation(s)
| | - Corey B Puryear
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA
| | | | - Sharan Srinivasan
- Praxis Precision Medicines, Boston, Massachusetts, 02110, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | - Ming-Kai Pan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, 10051, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, 10617, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, 10002, Taiwan.,Cerebellar Research Center, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, 64041, Taiwan
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, New York, 10032, USA.,Initiative for Columbia Ataxia and Tremor, Columbia University, New York, New York, 10032, USA
| |
Collapse
|
10
|
Wang C, Xu Z, Qiu X, Wei Y, Peralta AA, Yazdi MD, Jin T, Li W, Just A, Heiss J, Hou L, Zheng Y, Coull BA, Kosheleva A, Sparrow D, Amarasiriwardena C, Wright RO, Baccarelli AA, Schwartz JD. Epigenome-wide DNA methylation in leukocytes and toenail metals: The normative aging study. ENVIRONMENTAL RESEARCH 2023; 217:114797. [PMID: 36379232 PMCID: PMC9825663 DOI: 10.1016/j.envres.2022.114797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Environmental metal exposures have been associated with multiple deleterious health endpoints. DNA methylation (DNAm) may provide insight into the mechanisms underlying these relationships. Toenail metals are non-invasive biomarkers, reflecting a medium-term time exposure window. OBJECTIVES This study examined variation in leukocyte DNAm and toenail arsenic (As), cadmium (Cd), lead (Pb), manganese (Mn), and mercury (Hg) among elderly men in the Normative Aging Study, a longitudinal cohort. METHODS We repeatedly collected samples of blood and toenail clippings. We measured DNAm in leukocytes with the Illumina HumanMethylation450 K BeadChip. We first performed median regression to evaluate the effects of each individual toenail metal on DNAm at three levels: individual cytosine-phosphate-guanine (CpG) sites, regions, and pathways. Then, we applied a Bayesian kernel machine regression (BKMR) to assess the joint and individual effects of metal mixtures on DNAm. Significant CpGs were identified using a multiple testing correction based on the independent degrees of freedom approach for correlated outcomes. The approach considers the effective degrees of freedom in the DNAm data using the principal components that explain >95% variation of the data. RESULTS We included 564 subjects (754 visits) between 1999 and 2013. The numbers of significantly differentially methylated CpG sites, regions, and pathways varied by metals. For example, we found six significant pathways for As, three for Cd, and one for Mn. The As-associated pathways were associated with cancer (e.g., skin cancer) and cardiovascular disease, whereas the Cd-associated pathways were related to lung cancer. Metal mixtures were also associated with 47 significant CpG sites, as well as pathways, mainly related to cancer and cardiovascular disease. CONCLUSIONS This study provides an approach to understanding the potential epigenetic mechanisms underlying observed relations between toenail metals and adverse health endpoints.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Xinye Qiu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yaguang Wei
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Adjani A Peralta
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Mahdieh Danesh Yazdi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Tingfan Jin
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Wenyuan Li
- School of Public Health and Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Allan Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan Heiss
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Brent A Coull
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Anna Kosheleva
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - David Sparrow
- VA Normative Aging Study, VA Boston Healthcare System, Boston, MA 02130, USA; Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Chitra Amarasiriwardena
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY 10032, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
11
|
Mustafá ER, McCarthy CI, Portales AE, Cordisco Gonzalez S, Rodríguez SS, Raingo J. Constitutive activity of the dopamine (D 5 ) receptor, highly expressed in CA1 hippocampal neurons, selectively reduces Ca V 3.2 and Ca V 3.3 currents. Br J Pharmacol 2022; 180:1210-1231. [PMID: 36480023 DOI: 10.1111/bph.16006] [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: 06/07/2022] [Revised: 10/31/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE CaV 3.1-3 currents differentially contribute to neuronal firing patterns. CaV 3 are regulated by G protein-coupled receptors (GPCRs) activity, but information about CaV 3 as targets of the constitutive activity of GPCRs is scarce. We investigate the impact of D5 recpetor constitutive activity, a GPCR with high levels of basal activity, on CaV 3 functionality. D5 recpetor and CaV 3 are expressed in the hippocampus and have been independently linked to pathophysiological states associated with epilepsy. EXPERIMENTAL APPROACH Our study models were HEK293T cells heterologously expressing D1 or D5 receptor and CaV 3.1-3, and mouse brain slices containing the hippocampus. We used chlorpromazine (D1 /D5 inverse agonist) and a D5 receptor mutant lacking constitutive activity as experimental tools. We measured CaV 3 currents and excitability parameters using the patch-clamp technique. We completed our study with computational modelling and imaging technique. KEY RESULTS We found a higher sensitivity to TTA-P2 (CaV 3 blocker) in CA1 pyramidal neurons obtained from chlorpromazine-treated animals compared with vehicle-treated animals. We found that CaV 3.2 and CaV 3.3-but not CaV 3.1-are targets of D5 receptor constitutive activity in HEK293T cells. Finally, we found an increased firing rate in CA1 pyramidal neurons from chlorpromazine-treated animals in comparison with vehicle-treated animals. Similar changes in firing rate were observed on a neuronal model with controlled CaV 3 currents levels. CONCLUSIONS AND IMPLICATIONS Native hippocampal CaV 3 and recombinant CaV 3.2-3 are sensitive to D5 receptor constitutive activity. Manipulation of D5 receptor constitutive activity could be a valuable strategy to control neuronal excitability, especially in exacerbated conditions such as epilepsy.
Collapse
Affiliation(s)
- Emilio Román Mustafá
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| | - Clara Inés McCarthy
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| | - Andrea Estefanía Portales
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| | - Santiago Cordisco Gonzalez
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| | - Silvia Susana Rodríguez
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| | - Jesica Raingo
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], Buenos Aires, Argentina
| |
Collapse
|
12
|
Baez-Nieto D, Allen A, Akers-Campbell S, Yang L, Budnik N, Pupo A, Shin YC, Genovese G, Liao M, Pérez-Palma E, Heyne H, Lal D, Lipscombe D, Pan JQ. Analysing an allelic series of rare missense variants of CACNA1I in a Swedish schizophrenia cohort. Brain 2022; 145:1839-1853. [PMID: 34919654 PMCID: PMC9166571 DOI: 10.1093/brain/awab443] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/23/2021] [Accepted: 11/11/2021] [Indexed: 11/14/2022] Open
Abstract
CACNA1I is implicated in the susceptibility to schizophrenia by large-scale genetic association studies of single nucleotide polymorphisms. However, the channelopathy of CACNA1I in schizophrenia is unknown. CACNA1I encodes CaV3.3, a neuronal voltage-gated calcium channel that underlies a subtype of T-type current that is important for neuronal excitability in the thalamic reticular nucleus and other regions of the brain. Here, we present an extensive functional characterization of 57 naturally occurring rare and common missense variants of CACNA1I derived from a Swedish schizophrenia cohort of more than 10 000 individuals. Our analysis of this allelic series of coding CACNA1I variants revealed that reduced CaV3.3 channel current density was the dominant phenotype associated with rare CACNA1I coding alleles derived from control subjects, whereas rare CACNA1I alleles from schizophrenia patients encoded CaV3.3 channels with altered responses to voltages. CACNA1I variants associated with altered current density primarily impact the ionic channel pore and those associated with altered responses to voltage impact the voltage-sensing domain. CaV3.3 variants associated with altered voltage dependence of the CaV3.3 channel and those associated with peak current density deficits were significantly segregated across affected and unaffected groups (Fisher's exact test, P = 0.034). Our results, together with recent data from the SCHEMA (Schizophrenia Exome Sequencing Meta-Analysis) cohort, suggest that reduced CaV3.3 function may protect against schizophrenia risk in rare cases. We subsequently modelled the effect of the biophysical properties of CaV3.3 channel variants on thalamic reticular nucleus excitability and found that compared with common variants, ultrarare CaV3.3-coding variants derived from control subjects significantly decreased thalamic reticular nucleus excitability (P = 0.011). When all rare variants were analysed, there was a non-significant trend between variants that reduced thalamic reticular nucleus excitability and variants that either had no effect or increased thalamic reticular nucleus excitability across disease status. Taken together, the results of our functional analysis of an allelic series of >50 CACNA1I variants in a schizophrenia cohort reveal that loss of function of CaV3.3 is a molecular phenotype associated with reduced disease risk burden, and our approach may serve as a template strategy for channelopathies in polygenic disorders.
Collapse
Affiliation(s)
- David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Andrew Allen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Seth Akers-Campbell
- Carney Institute for Brain Science & Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nikita Budnik
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Amaury Pupo
- Department of Biology, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Young-Cheul Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Maofu Liao
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Eduardo Pérez-Palma
- Genomic Medicine Institute, Lerner Research institute, Cleveland Clinic, OH 44195, USA
- Centro de Genética y Genómica, Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Chile
| | - Henrike Heyne
- Genomic Medicine, Hasso Plattner Institute, Potsdam, 14482, Germany
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research institute, Cleveland Clinic, OH 44195, USA
- Cologne Center for Genomics, University of Cologne, Cologne 50931, Germany
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Diane Lipscombe
- Carney Institute for Brain Science & Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
13
|
Erdogmus S, Concepcion AR, Yamashita M, Sidhu I, Tao AY, Li W, Rocha PP, Huang B, Garippa R, Lee B, Lee A, Hell JW, Lewis RS, Prakriya M, Feske S. Cavβ1 regulates T cell expansion and apoptosis independently of voltage-gated Ca 2+ channel function. Nat Commun 2022; 13:2033. [PMID: 35440113 PMCID: PMC9018955 DOI: 10.1038/s41467-022-29725-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
Abstract
TCR stimulation triggers Ca2+ signals that are critical for T cell function and immunity. Several pore-forming α and auxiliary β subunits of voltage-gated Ca2+ channels (VGCC) were reported in T cells, but their mechanism of activation remains elusive and their contribution to Ca2+ signaling in T cells is controversial. We here identify CaVβ1, encoded by Cacnb1, as a regulator of T cell function. Cacnb1 deletion enhances apoptosis and impairs the clonal expansion of T cells after lymphocytic choriomeningitis virus (LCMV) infection. By contrast, Cacnb1 is dispensable for T cell proliferation, cytokine production and Ca2+ signaling. Using patch clamp electrophysiology and Ca2+ recordings, we are unable to detect voltage-gated Ca2+ currents or Ca2+ influx in human and mouse T cells upon depolarization with or without prior TCR stimulation. mRNAs of several VGCC α1 subunits are detectable in human (CaV3.3, CaV3.2) and mouse (CaV2.1) T cells, but they lack transcription of many 5' exons, likely resulting in N-terminally truncated and non-functional proteins. Our findings demonstrate that although CaVβ1 regulates T cell function, these effects are independent of VGCC channel activity.
Collapse
Affiliation(s)
- Serap Erdogmus
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Axel R Concepcion
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Megumi Yamashita
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Ikjot Sidhu
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Anthony Y Tao
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Wenyi Li
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Pedro P Rocha
- Unit on Genome Structure and Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- National Cancer Institute, NIH, Bethesda, MD, USA
| | - Bonnie Huang
- National Institute of Allergy and Infectious Disease, Bethesda, MD, USA
- National Human Genome Research Institute, Bethesda, MD, USA
| | - Ralph Garippa
- Department of Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Boram Lee
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, Austin, TX, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Richard S Lewis
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
| | - Stefan Feske
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Zhang YL, Moran SP, Allen A, Baez-Nieto D, Xu Q, Wang LA, Martenis WE, Sacher JR, Gale JP, Weïwer M, Wagner FF, Pan JQ. Novel Fluorescence-Based High-Throughput FLIPR Assay Utilizing Membrane-Tethered Genetic Calcium Sensors to Identify T-Type Calcium Channel Modulators. ACS Pharmacol Transl Sci 2022; 5:156-168. [PMID: 35311021 PMCID: PMC8923061 DOI: 10.1021/acsptsci.1c00233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Indexed: 11/28/2022]
Abstract
T-type voltage-gated Ca2+ channels have been implicated in many human disorders, and there has been increasing interest in developing highly selective and potent T-type Ca2+ channel modulators for potential clinical use. However, the unique biophysical properties of T-type Ca2+ channels are not conducive for developing high-throughput screening (HTS) assays to identify modulators, particularly potentiators. To illustrate, T-type Ca2+ channels are largely inactivated and unable to open to allow Ca2+ influx at -25 mV, the typical resting membrane potential of the cell lines commonly used in cellular screening assays. To address this issue, we developed cell lines that express Kir2.3 channels to hyperpolarize the membrane potential to -70 mV, thus allowing T-type channels to return to their resting state where they can be subsequently activated by membrane depolarization in the presence of extracellular KCl. Furthermore, to simplify the HTS assay and to reduce reagent cost, we stably expressed a membrane-tethered genetic calcium sensor, GCaMP6s-CAAX, that displays superior signal to the background compared to the untethered GCaMP6s or the synthetic Ca2+ sensor Fluo-4AM. Here, we describe a novel GCaMP6s-CAAX-based calcium assay utilizing a high-throughput fluorometric imaging plate reader (Molecular Devices, Sunnyvale, CA) format that can identify both activators and inhibitors of T-type Ca2+ channels. Lastly, we demonstrate the utility of this novel fluorescence-based assay to evaluate the activities of two distinct G-protein-coupled receptors, thus expanding the use of GCaMP6s-CAAX to a wide range of applications relevant for developing cellular assays in drug discovery.
Collapse
Affiliation(s)
- Yan-Ling Zhang
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sean P. Moran
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andrew Allen
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - David Baez-Nieto
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Qihong Xu
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Lei A. Wang
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - William E. Martenis
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Joshua R. Sacher
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jennifer P. Gale
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Michel Weïwer
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Florence F. Wagner
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jen Q. Pan
- Stanley Center for Psychiatric
Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
15
|
Klemz A, Wildner F, Tütüncü E, Gerevich Z. Regulation of Hippocampal Gamma Oscillations by Modulation of Intrinsic Neuronal Excitability. Front Neural Circuits 2022; 15:778022. [PMID: 35177966 PMCID: PMC8845518 DOI: 10.3389/fncir.2021.778022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Ion channels activated around the subthreshold membrane potential determine the likelihood of neuronal firing in response to synaptic inputs, a process described as intrinsic neuronal excitability. Long-term plasticity of chemical synaptic transmission is traditionally considered the main cellular mechanism of information storage in the brain; however, voltage- and calcium-activated channels modulating the inputs or outputs of neurons are also subjects of plastic changes and play a major role in learning and memory formation. Gamma oscillations are associated with numerous higher cognitive functions such as learning and memory, but our knowledge of their dependence on intrinsic plasticity is by far limited. Here we investigated the roles of potassium and calcium channels activated at near subthreshold membrane potentials in cholinergically induced persistent gamma oscillations measured in the CA3 area of rat hippocampal slices. Among potassium channels, which are responsible for the afterhyperpolarization in CA3 pyramidal cells, we found that blockers of SK (KCa2) and KV7.2/7.3 (KCNQ2/3), but not the BK (KCa1.1) and IK (KCa3.1) channels, increased the power of gamma oscillations. On the contrary, activators of these channels had an attenuating effect without affecting the frequency. Pharmacological blockade of the low voltage-activated T-type calcium channels (CaV3.1–3.3) reduced gamma power and increased the oscillation peak frequency. Enhancement of these channels also inhibited the peak power without altering the frequency of the oscillations. The presented data suggest that voltage- and calcium-activated ion channels involved in intrinsic excitability strongly regulate the power of hippocampal gamma oscillations. Targeting these channels could represent a valuable pharmacological strategy against cognitive impairment.
Collapse
|
16
|
Maksemous N, Blayney CD, Sutherland HG, Smith RA, Lea RA, Tran KN, Ibrahim O, McArthur JR, Haupt LM, Cader MZ, Finol-Urdaneta RK, Adams DJ, Griffiths LR. Investigation of CACNA1I Cav3.3 Dysfunction in Hemiplegic Migraine. Front Mol Neurosci 2022; 15:892820. [PMID: 35928792 PMCID: PMC9345121 DOI: 10.3389/fnmol.2022.892820] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/13/2022] [Indexed: 01/12/2023] Open
Abstract
Familial hemiplegic migraine (FHM) is a severe neurogenetic disorder for which three causal genes, CACNA1A, SCN1A, and ATP1A2, have been implicated. However, more than 80% of referred diagnostic cases of hemiplegic migraine (HM) are negative for exonic mutations in these known FHM genes, suggesting the involvement of other genes. Using whole-exome sequencing data from 187 mutation-negative HM cases, we identified rare variants in the CACNA1I gene encoding the T-type calcium channel Cav3.3. Burden testing of CACNA1I variants showed a statistically significant increase in allelic burden in the HM case group compared to gnomAD (OR = 2.30, P = 0.00005) and the UK Biobank (OR = 2.32, P = 0.0004) databases. Dysfunction in T-type calcium channels, including Cav3.3, has been implicated in a range of neurological conditions, suggesting a potential role in HM. Using patch-clamp electrophysiology, we compared the biophysical properties of five Cav3.3 variants (p.R111G, p.M128L, p.D302G, p.R307H, and p.Q1158H) to wild-type (WT) channels expressed in HEK293T cells. We observed numerous functional alterations across the channels with Cav3.3-Q1158H showing the greatest differences compared to WT channels, including reduced current density, right-shifted voltage dependence of activation and inactivation, and slower current kinetics. Interestingly, we also found significant differences in the conductance properties exhibited by the Cav3.3-R307H and -Q1158H variants compared to WT channels under conditions of acidosis and alkalosis. In light of these data, we suggest that rare variants in CACNA1I may contribute to HM etiology.
Collapse
Affiliation(s)
- Neven Maksemous
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Claire D Blayney
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Heidi G Sutherland
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Robert A Smith
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rod A Lea
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kim Ngan Tran
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Omar Ibrahim
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Larisa M Haupt
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - M Zameel Cader
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, The Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Jiang Y, Patton MH, Zakharenko SS. A Case for Thalamic Mechanisms of Schizophrenia: Perspective From Modeling 22q11.2 Deletion Syndrome. Front Neural Circuits 2021; 15:769969. [PMID: 34955759 PMCID: PMC8693383 DOI: 10.3389/fncir.2021.769969] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a severe, chronic psychiatric disorder that devastates the lives of millions of people worldwide. The disease is characterized by a constellation of symptoms, ranging from cognitive deficits, to social withdrawal, to hallucinations. Despite decades of research, our understanding of the neurobiology of the disease, specifically the neural circuits underlying schizophrenia symptoms, is still in the early stages. Consequently, the development of therapies continues to be stagnant, and overall prognosis is poor. The main obstacle to improving the treatment of schizophrenia is its multicausal, polygenic etiology, which is difficult to model. Clinical observations and the emergence of preclinical models of rare but well-defined genomic lesions that confer substantial risk of schizophrenia (e.g., 22q11.2 microdeletion) have highlighted the role of the thalamus in the disease. Here we review the literature on the molecular, cellular, and circuitry findings in schizophrenia and discuss the leading theories in the field, which point to abnormalities within the thalamus as potential pathogenic mechanisms of schizophrenia. We posit that synaptic dysfunction and oscillatory abnormalities in neural circuits involving projections from and within the thalamus, with a focus on the thalamocortical circuits, may underlie the psychotic (and possibly other) symptoms of schizophrenia.
Collapse
Affiliation(s)
| | | | - Stanislav S. Zakharenko
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
18
|
Abstract
Sleep disturbances are commonly observed in schizophrenia, including in chronic, early-course, and first-episode patients. This has generated considerable interest, both in clinical and research endeavors, in characterizing the relationship between disturbed sleep and schizophrenia. Sleep features can be objectively assessed with EEG recordings. Traditionally, EEG studies have focused on sleep architecture, which includes non-REM and REM sleep stages. More recently, numerous studies have investigated alterations in sleep-specific rhythms, including EEG oscillations, such as sleep spindles and slow waves, in individuals with schizophrenia compared with control subjects. In this article, the author reviews state-of-the-art evidence of disturbed sleep in schizophrenia, starting from the relationship between sleep disturbances and clinical symptoms. First, the author presents studies demonstrating abnormalities in sleep architecture and sleep-oscillatory rhythms in schizophrenia and related psychotic disorders, with an emphasis on recent work demonstrating sleep spindles and slow-wave deficits in early-course and first-episode schizophrenia. Next, the author shows how these sleep abnormalities relate to the cognitive impairments in patients diagnosed with schizophrenia and point to dysfunctions in underlying thalamocortical circuits, Ca+ channel activity, and GABA-glutamate neurotransmission. Finally, the author discusses some of the next steps needed to further establish the role of altered sleep in schizophrenia, including the need to investigate sleep abnormalities across the psychotic spectrum and to establish their relationship with circadian disturbances, which in turn will contribute to the development of novel sleep-informed treatment interventions.
Collapse
Affiliation(s)
- Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh School of Medicine Pittsburgh, PA, 15213
| |
Collapse
|
19
|
El Ghaleb Y, Schneeberger PE, Fernández-Quintero ML, Geisler SM, Pelizzari S, Polstra AM, van Hagen JM, Denecke J, Campiglio M, Liedl KR, Stevens CA, Person RE, Rentas S, Marsh ED, Conlin LK, Tuluc P, Kutsche K, Flucher BE. CACNA1I gain-of-function mutations differentially affect channel gating and cause neurodevelopmental disorders. Brain 2021; 144:2092-2106. [PMID: 33704440 PMCID: PMC8422349 DOI: 10.1093/brain/awab101] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
T-type calcium channels (Cav3.1 to Cav3.3) regulate low-threshold calcium spikes, burst firing and rhythmic oscillations of neurons and are involved in sensory processing, sleep, and hormone and neurotransmitter release. Here, we examined four heterozygous missense variants in CACNA1I, encoding the Cav3.3 channel, in patients with variable neurodevelopmental phenotypes. The p.(Ile860Met) variant, affecting a residue in the putative channel gate at the cytoplasmic end of the IIS6 segment, was identified in three family members with variable cognitive impairment. The de novo p.(Ile860Asn) variant, changing the same amino acid residue, was detected in a patient with severe developmental delay and seizures. In two additional individuals with global developmental delay, hypotonia, and epilepsy, the variants p.(Ile1306Thr) and p.(Met1425Ile), substituting residues at the cytoplasmic ends of IIIS5 and IIIS6, respectively, were found. Because structure modelling indicated that the amino acid substitutions differentially affect the mobility of the channel gate, we analysed possible effects on Cav3.3 channel function using patch-clamp analysis in HEK293T cells. The mutations resulted in slowed kinetics of current activation, inactivation, and deactivation, and in hyperpolarizing shifts of the voltage-dependence of activation and inactivation, with Cav3.3-I860N showing the strongest and Cav3.3-I860M the weakest effect. Structure modelling suggests that by introducing stabilizing hydrogen bonds the mutations slow the kinetics of the channel gate and cause the gain-of-function effect in Cav3.3 channels. The gating defects left-shifted and increased the window currents, resulting in increased calcium influx during repetitive action potentials and even at resting membrane potentials. Thus, calcium toxicity in neurons expressing the Cav3.3 variants is one likely cause of the neurodevelopmental phenotype. Computer modelling of thalamic reticular nuclei neurons indicated that the altered gating properties of the Cav3.3 disease variants lower the threshold and increase the duration and frequency of action potential firing. Expressing the Cav3.3-I860N/M mutants in mouse chromaffin cells shifted the mode of firing from low-threshold spikes and rebound burst firing with wild-type Cav3.3 to slow oscillations with Cav3.3-I860N and an intermediate firing mode with Cav3.3-I860M, respectively. Such neuronal hyper-excitability could explain seizures in the patient with the p.(Ile860Asn) mutation. Thus, our study implicates CACNA1I gain-of-function mutations in neurodevelopmental disorders, with a phenotypic spectrum ranging from borderline intellectual functioning to a severe neurodevelopmental disorder with epilepsy.
Collapse
Affiliation(s)
- Yousra El Ghaleb
- Institute of Physiology, Medical University Innsbruck, Innsbruck 6020, Austria
| | - Pauline E Schneeberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Monica L Fernández-Quintero
- Institute of Physiology, Medical University Innsbruck, Innsbruck 6020, Austria
- Institute of Theoretical Chemistry, University of Innsbruck, Innsbruck 6020, Austria
| | - Stefanie M Geisler
- Department of Pharmacology, University of Innsbruck, Innsbruck 6020, Austria
| | - Simone Pelizzari
- Institute of Physiology, Medical University Innsbruck, Innsbruck 6020, Austria
| | - Abeltje M Polstra
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1012 WX, The Netherlands
| | - Johanna M van Hagen
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1012 WX, The Netherlands
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, Innsbruck 6020, Austria
| | - Klaus R Liedl
- Institute of Theoretical Chemistry, University of Innsbruck, Innsbruck 6020, Austria
| | - Cathy A Stevens
- Department of Pediatrics, University of Tennessee College of Medicine, Chattanooga, TN 37403, USA
| | | | - Stefan Rentas
- Division of Genomic Diagnostics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eric D Marsh
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Laura K Conlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Petronel Tuluc
- Department of Pharmacology, University of Innsbruck, Innsbruck 6020, Austria
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Bernhard E Flucher
- Institute of Physiology, Medical University Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
20
|
Steullet P. Thalamus-related anomalies as candidate mechanism-based biomarkers for psychosis. Schizophr Res 2020; 226:147-157. [PMID: 31147286 DOI: 10.1016/j.schres.2019.05.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 02/08/2023]
Abstract
Identification of reliable biomarkers of prognosis in subjects with high risk to psychosis is an essential step to improve care and treatment of this population of help-seekers. Longitudinal studies highlight some clinical criteria, cognitive deficits, patterns of gray matter alterations and profiles of blood metabolites that provide some levels of prediction regarding the conversion to psychosis. Further effort is warranted to validate these results and implement these types of approaches in clinical settings. Such biomarkers may however fall short in entangling the biological mechanisms underlying the disease progression, an essential step in the development of novel therapies. Circuit-based approaches, which map on well-identified cerebral functions, could meet these needs. Converging evidence indicates that thalamus abnormalities are central to schizophrenia pathophysiology, contributing to clinical symptoms, cognitive and sensory deficits. This review highlights the various thalamus-related anomalies reported in individuals with genetic risks and in the different phases of the disorder, from prodromal to chronic stages. Several anomalies are potent endophenotypes, while others exist in clinical high-risk subjects and worsen in those who convert to full psychosis. Aberrant functional coupling between thalamus and cortex, low glutamate content and readouts from resting EEG carry predictive values for transition to psychosis or functional outcome. In this context, thalamus-related anomalies represent a valuable entry point to tackle circuit-based alterations associated with the emergence of psychosis. This review also proposes that longitudinal surveys of neuroimaging, EEG readouts associated with circuits encompassing the mediodorsal, pulvinar in high-risk individuals could unveil biological mechanisms contributing to this psychiatric disorder.
Collapse
Affiliation(s)
- Pascal Steullet
- Center of Psychiatric Neuroscience, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Site de Cery, 1008 Prilly-Lausanne, Switzerland.
| |
Collapse
|
21
|
Mealer RG, Williams SE, Daly MJ, Scolnick EM, Cummings RD, Smoller JW. Glycobiology and schizophrenia: a biological hypothesis emerging from genomic research. Mol Psychiatry 2020; 25:3129-3139. [PMID: 32377000 PMCID: PMC8081046 DOI: 10.1038/s41380-020-0753-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022]
Abstract
Advances in genomics are opening new windows into the biology of schizophrenia. Though common variants individually have small effects on disease risk, GWAS provide a powerful opportunity to explore pathways and mechanisms contributing to pathophysiology. Here, we highlight an underappreciated biological theme emerging from GWAS: the role of glycosylation in schizophrenia. The strongest coding variant in schizophrenia GWAS is a missense mutation in the manganese transporter SLC39A8, which is associated with altered glycosylation patterns in humans. Furthermore, variants near several genes encoding glycosylation enzymes are unambiguously associated with schizophrenia: FUT9, MAN2A1, TMTC1, GALNT10, and B3GAT1. Here, we summarize the known biological functions, target substrates, and expression patterns of these enzymes as a primer for future studies. We also highlight a subset of schizophrenia-associated proteins critically modified by glycosylation including glutamate receptors, voltage-gated calcium channels, the dopamine D2 receptor, and complement glycoproteins. We hypothesize that common genetic variants alter brain glycosylation and play a fundamental role in the development of schizophrenia. Leveraging these findings will advance our mechanistic understanding of disease and may provide novel avenues for treatment development.
Collapse
Affiliation(s)
- Robert G. Mealer
- Massachusetts General Hospital, Department of Psychiatry.,The Stanley Center for Psychiatric Research at Broad Institute.,Department of Surgery, Beth Israel Deaconess Medical Center. Harvard Medical School, Boston MA.,Corresponding Author: Robert Gene Mealer, M.D., Ph.D., Richard B. Simches Research Center, 185 Cambridge St, 6th Floor, Boston, MA 02114, Tel: +1 (617) 724-9076,
| | - Sarah E. Williams
- Massachusetts General Hospital, Department of Psychiatry.,Department of Surgery, Beth Israel Deaconess Medical Center. Harvard Medical School, Boston MA
| | - Mark J. Daly
- Massachusetts General Hospital, Department of Psychiatry.,The Stanley Center for Psychiatric Research at Broad Institute
| | - Edward M. Scolnick
- Massachusetts General Hospital, Department of Psychiatry.,The Stanley Center for Psychiatric Research at Broad Institute
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center. Harvard Medical School, Boston MA
| | - Jordan W. Smoller
- Massachusetts General Hospital, Department of Psychiatry.,The Stanley Center for Psychiatric Research at Broad Institute
| |
Collapse
|
22
|
Heyne HO, Baez-Nieto D, Iqbal S, Palmer DS, Brunklaus A, May P, Johannesen KM, Lauxmann S, Lemke JR, Møller RS, Pérez-Palma E, Scholl UI, Syrbe S, Lerche H, Lal D, Campbell AJ, Wang HR, Pan J, Daly MJ. Predicting functional effects of missense variants in voltage-gated sodium and calcium channels. Sci Transl Med 2020; 12:eaay6848. [PMID: 32801145 DOI: 10.1126/scitranslmed.aay6848] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/20/2019] [Accepted: 07/22/2020] [Indexed: 12/30/2022]
Abstract
Malfunctions of voltage-gated sodium and calcium channels (encoded by SCNxA and CACNA1x family genes, respectively) have been associated with severe neurologic, psychiatric, cardiac, and other diseases. Altered channel activity is frequently grouped into gain or loss of ion channel function (GOF or LOF, respectively) that often corresponds not only to clinical disease manifestations but also to differences in drug response. Experimental studies of channel function are therefore important, but laborious and usually focus only on a few variants at a time. On the basis of known gene-disease mechanisms of 19 different diseases, we inferred LOF (n = 518) and GOF (n = 309) likely pathogenic variants from the disease phenotypes of variant carriers. By training a machine learning model on sequence- and structure-based features, we predicted LOF or GOF effects [area under the receiver operating characteristics curve (ROC) = 0.85] of likely pathogenic missense variants. Our LOF versus GOF prediction corresponded to molecular LOF versus GOF effects for 87 functionally tested variants in SCN1/2/8A and CACNA1I (ROC = 0.73) and was validated in exome-wide data from 21,703 cases and 128,957 controls. We showed respective regional clustering of inferred LOF and GOF nucleotide variants across the alignment of the entire gene family, suggesting shared pathomechanisms in the SCNxA/CACNA1x family genes.
Collapse
Affiliation(s)
- Henrike O Heyne
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 5WR36M Helsinki, Finland
| | - David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sumaiya Iqbal
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Duncan S Palmer
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andreas Brunklaus
- Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow G51 4TF, UK
- School of Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, Belvaux, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Katrine M Johannesen
- Department of Epilepsy Genetics and Personalized Treatment, Danish Epilepsy Centre, 4293 Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Treatment, Danish Epilepsy Centre, 4293 Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Eduardo Pérez-Palma
- Cologne Center for Genomics (CCG), University of Cologne, 50923, Germany
- Genomic Medicine Institute, Lemer Research Institute Cleveland Clinic, OH G92J47, USA
| | - Ute I Scholl
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology and Medical Intensive Care and BIH Center for Regenerative Therapies, 10178 Berlin, Germany
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Center for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Dennis Lal
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Cologne Center for Genomics (CCG), University of Cologne, 50923, Germany
- Genomic Medicine Institute, Lemer Research Institute Cleveland Clinic, OH G92J47, USA
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH G92J47, USA
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hao-Ran Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jen Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 5WR36M Helsinki, Finland
| |
Collapse
|
23
|
A single psychotomimetic dose of ketamine decreases thalamocortical spindles and delta oscillations in the sedated rat. Schizophr Res 2020; 222:362-374. [PMID: 32507548 DOI: 10.1016/j.schres.2020.04.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/18/2020] [Accepted: 04/19/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND In patients with psychotic disorders, sleep spindles are reduced, supporting the hypothesis that the thalamus and glutamate receptors play a crucial etio-pathophysiological role, whose underlying mechanisms remain unknown. We hypothesized that a reduced function of NMDA receptors is involved in the spindle deficit observed in schizophrenia. METHODS An electrophysiological multisite cell-to-network exploration was used to investigate, in pentobarbital-sedated rats, the effects of a single psychotomimetic dose of the NMDA glutamate receptor antagonist ketamine in the sensorimotor and associative/cognitive thalamocortical (TC) systems. RESULTS Under the control condition, spontaneously-occurring spindles (intra-frequency: 10-16 waves/s) and delta-frequency (1-4 Hz) oscillations were recorded in the frontoparietal cortical EEG, in thalamic extracellular recordings, in dual juxtacellularly recorded GABAergic thalamic reticular nucleus (TRN) and glutamatergic TC neurons, and in intracellularly recorded TC neurons. The TRN cells rhythmically exhibited robust high-frequency bursts of action potentials (7 to 15 APs at 200-700 Hz). A single administration of low-dose ketamine fleetingly reduced TC spindles and delta oscillations, amplified ongoing gamma-(30-80 Hz) and higher-frequency oscillations, and switched the firing pattern of both TC and TRN neurons from a burst mode to a single AP mode. Furthermore, ketamine strengthened the gamma-frequency band TRN-TC connectivity. The antipsychotic clozapine consistently prevented the ketamine effects on spindles, delta- and gamma-/higher-frequency TC oscillations. CONCLUSION The present findings support the hypothesis that NMDA receptor hypofunction is involved in the reduction in sleep spindles and delta oscillations. The ketamine-induced swift conversion of ongoing TC-TRN activities may have involved at least both the ascending reticular activating system and the corticothalamic pathway.
Collapse
|
24
|
Lory P, Nicole S, Monteil A. Neuronal Cav3 channelopathies: recent progress and perspectives. Pflugers Arch 2020; 472:831-844. [PMID: 32638069 PMCID: PMC7351805 DOI: 10.1007/s00424-020-02429-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
T-type, low-voltage activated, calcium channels, now designated Cav3 channels, are involved in a wide variety of physiological functions, especially in nervous systems. Their unique electrophysiological properties allow them to finely regulate neuronal excitability and to contribute to sensory processing, sleep, and hormone and neurotransmitter release. In the last two decades, genetic studies, including exploration of knock-out mouse models, have greatly contributed to elucidate the role of Cav3 channels in normal physiology, their regulation, and their implication in diseases. Mutations in genes encoding Cav3 channels (CACNA1G, CACNA1H, and CACNA1I) have been linked to a variety of neurodevelopmental, neurological, and psychiatric diseases designated here as neuronal Cav3 channelopathies. In this review, we describe and discuss the clinical findings and supporting in vitro and in vivo studies of the mutant channels, with a focus on de novo, gain-of-function missense mutations recently discovered in CACNA1G and CACNA1H. Overall, the studies of the Cav3 channelopathies help deciphering the pathogenic mechanisms of corresponding diseases and better delineate the properties and physiological roles Cav3 channels.
Collapse
Affiliation(s)
- Philippe Lory
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France. .,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France.
| | - Sophie Nicole
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| |
Collapse
|
25
|
Manoach DS, Mylonas D, Baxter B. Targeting sleep oscillations to improve memory in schizophrenia. Schizophr Res 2020; 221:63-70. [PMID: 32014359 PMCID: PMC7316628 DOI: 10.1016/j.schres.2020.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/26/2022]
Abstract
Although schizophrenia is defined by waking phenomena, a growing literature documents a deficit in sleep spindles, a defining oscillation of stage 2 non-rapid eye movement sleep. Compelling evidence supports an important role for spindles in cognition, and particularly memory. In schizophrenia, although the spindle deficit correlates with impaired sleep-dependent memory consolidation, recent clinical trials find that increasing spindles does not improve memory. This may reflect that sleep-dependent memory consolidation relies not on spindles alone, but also on their precise temporal coordination with cortical slow oscillations and hippocampal sharp-wave ripples. Consequently, interventions to improve memory in schizophrenia must not only increase spindles, but also preserve or enhance slow oscillations, hippocampal ripples and their temporal relations. Because hippocampal ripples and the activity of the thalamic spindle generator are difficult to measure noninvasively, screening potential interventions requires complementary animal and human studies. In this review we (i) propose that sleep oscillations are novel pathophysiological targets for therapy to improve cognition in schizophrenia; (ii) summarize our understanding of how these oscillations interact to consolidate memory; (iii) suggest that a systems neuroscience strategy is essential to selecting and evaluating effective treatments, and illustrate this with findings from clinical trials; and (iv) selectively review the interventional literature relevant to sleep and cognition, covering both pharmacological and noninvasive brain stimulation approaches. We conclude that coordinated sleep oscillations are promising targets for improving cognition in schizophrenia and that effective therapies will need to preserve or enhance sleep oscillatory dynamics and restore function at the network level.
Collapse
Affiliation(s)
- Dara S Manoach
- Department of Psychiatry Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | - Dimitrios Mylonas
- Department of Psychiatry Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bryan Baxter
- Department of Psychiatry Massachusetts General Hospital, Harvard Medical School, Boston, MA 02215, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
26
|
Rodenkirch C, Wang Q. Rapid and transient enhancement of thalamic information transmission induced by vagus nerve stimulation. J Neural Eng 2020; 17:026027. [PMID: 31935689 DOI: 10.1088/1741-2552/ab6b84] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Vagus nerve stimulation (VNS) has been FDA-approved as a long-term, therapeutic treatment for multiple disorders, including pharmacoresistant epilepsy and depression. Here we elucidate the short-term effects of VNS on sensory processing. APPROACH We employed an information theoretic approach to examine the effects of VNS on thalamocortical transmission of sensory-related information along the somatosensory pathway. MAIN RESULTS We found that VNS enhanced the selectivity of the response of thalamic neurons to specific kinetic features in the stimuli, resulting in a significant increase in the efficiency and rate of stimulus-related information conveyed by thalamic spikes. VNS-induced improvements in thalamic sensory processing coincided with a decrease in thalamic burst firing. Importantly, we found VNS-induced enhancement of sensory processing had a rapid onset and offset, completely disappearing one minute after cessation of VNS. The timescales of these effects indicate against an underlying mechanism involving long-term neuroplasticity. We found several patterns of VNS (tonic, standard duty-cycle, and fast duty-cycle) all induced similar improvements in sensory processing. Under closer inspection we noticed that due to the fast timescale of VNS effects on sensory processing, standard duty-cycle VNS induced a fluctuating sensory processing state which may be sub-optimal for perceptual behavior. Fast duty-cycle VNS and continuous, tonic VNS induced quantitatively similar improvements in thalamic information transmission as standard duty-cycle VNS without inducing a fluctuating thalamic state. Further, we found the strength of VNS-induced improvements in sensory processing increased monotonically with amplitude and frequency of VNS. SIGNIFICANCE These results demonstrate, for the first time, the feasibility of utilizing specific patterns of VNS to rapidly improve sensory processing and confirm fast duty-cycle and tonic patterns as optimal for this purpose, while showing standard duty-cycle VNS causes non-optimal fluctuations in thalamic state.
Collapse
Affiliation(s)
- Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, ET351, 500 W. 120th Street, New York, NY 10027, United States of America
| | | |
Collapse
|
27
|
Ghoshal A, Uygun DS, Yang L, McNally JM, Lopez-Huerta VG, Arias-Garcia MA, Baez-Nieto D, Allen A, Fitzgerald M, Choi S, Zhang Q, Hope JM, Yan K, Mao X, Nicholson TB, Imaizumi K, Fu Z, Feng G, Brown RE, Strecker RE, Purcell SM, Pan JQ. Effects of a patient-derived de novo coding alteration of CACNA1I in mice connect a schizophrenia risk gene with sleep spindle deficits. Transl Psychiatry 2020; 10:29. [PMID: 32066662 PMCID: PMC7026444 DOI: 10.1038/s41398-020-0685-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 10/25/2019] [Accepted: 11/06/2019] [Indexed: 11/09/2022] Open
Abstract
CACNA1I, a schizophrenia risk gene, encodes a subtype of voltage-gated T-type calcium channel CaV3.3. We previously reported that a patient-derived missense de novo mutation (R1346H) of CACNA1I impaired CaV3.3 channel function. Here, we generated CaV3.3-RH knock-in animals, along with mice lacking CaV3.3, to investigate the biological impact of R1346H (RH) variation. We found that RH mutation altered cellular excitability in the thalamic reticular nucleus (TRN), where CaV3.3 is abundantly expressed. Moreover, RH mutation produced marked deficits in sleep spindle occurrence and morphology throughout non-rapid eye movement (NREM) sleep, while CaV3.3 haploinsufficiency gave rise to largely normal spindles. Therefore, mice harboring the RH mutation provide a patient derived genetic model not only to dissect the spindle biology but also to evaluate the effects of pharmacological reagents in normalizing sleep spindle deficits. Importantly, our analyses highlighted the significance of characterizing individual spindles and strengthen the inferences we can make across species over sleep spindles. In conclusion, this study established a translational link between a genetic allele and spindle deficits during NREM observed in schizophrenia patients, representing a key step toward testing the hypothesis that normalizing spindles may be beneficial for schizophrenia patients.
Collapse
Affiliation(s)
- Ayan Ghoshal
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - David S. Uygun
- Department of Psychiatry, VA Boston Healthcare System & Harvard Medical School, Boston, MA USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - James M. McNally
- Department of Psychiatry, VA Boston Healthcare System & Harvard Medical School, Boston, MA USA
| | - Violeta G. Lopez-Huerta
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
- Present Address: Department of Neurodevelopment and Physiology, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mario A. Arias-Garcia
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
- Present Address: Department of Neurodevelopment and Physiology, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - Andrew Allen
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - Megan Fitzgerald
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - Soonwook Choi
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
| | - Qiangge Zhang
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
| | - Jen M. Hope
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - Karena Yan
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| | - Xiaohong Mao
- Novartis Institutes for BioMedical Research, 181 Mass Ave., Cambridge, MA 02139 USA
| | - Thomas B. Nicholson
- Novartis Institutes for BioMedical Research, 181 Mass Ave., Cambridge, MA 02139 USA
| | | | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
| | - Ritchie E. Brown
- Department of Psychiatry, VA Boston Healthcare System & Harvard Medical School, Boston, MA USA
| | - Robert E. Strecker
- Department of Psychiatry, VA Boston Healthcare System & Harvard Medical School, Boston, MA USA
| | - Shaun M. Purcell
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA USA
| |
Collapse
|
28
|
Weiss N, Zamponi GW. Genetic T-type calcium channelopathies. J Med Genet 2020; 57:1-10. [PMID: 31217264 PMCID: PMC6929700 DOI: 10.1136/jmedgenet-2019-106163] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/02/2019] [Accepted: 05/18/2019] [Indexed: 12/13/2022]
Abstract
T-type channels are low-voltage-activated calcium channels that contribute to a variety of cellular and physiological functions, including neuronal excitability, hormone and neurotransmitter release as well as developmental aspects. Several human conditions including epilepsy, autism spectrum disorders, schizophrenia, motor neuron disorders and aldosteronism have been traced to variations in genes encoding T-type channels. In this short review, we present the genetics of T-type channels with an emphasis on structure-function relationships and associated channelopathies.
Collapse
Affiliation(s)
- Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Praha, Czech Republic
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
29
|
Abstract
Sleep spindles are burstlike signals in the electroencephalogram (EEG) of the sleeping mammalian brain and electrical surface correlates of neuronal oscillations in thalamus. As one of the most inheritable sleep EEG signatures, sleep spindles probably reflect the strength and malleability of thalamocortical circuits that underlie individual cognitive profiles. We review the characteristics, organization, regulation, and origins of sleep spindles and their implication in non-rapid-eye-movement sleep (NREMS) and its functions, focusing on human and rodent. Spatially, sleep spindle-related neuronal activity appears on scales ranging from small thalamic circuits to functional cortical areas, and generates a cortical state favoring intracortical plasticity while limiting cortical output. Temporally, sleep spindles are discrete events, part of a continuous power band, and elements grouped on an infraslow time scale over which NREMS alternates between continuity and fragility. We synthesize diverse and seemingly unlinked functions of sleep spindles for sleep architecture, sensory processing, synaptic plasticity, memory formation, and cognitive abilities into a unifying sleep spindle concept, according to which sleep spindles 1) generate neural conditions of large-scale functional connectivity and plasticity that outlast their appearance as discrete EEG events, 2) appear preferentially in thalamic circuits engaged in learning and attention-based experience during wakefulness, and 3) enable a selective reactivation and routing of wake-instated neuronal traces between brain areas such as hippocampus and cortex. Their fine spatiotemporal organization reflects NREMS as a physiological state coordinated over brain and body and may indicate, if not anticipate and ultimately differentiate, pathologies in sleep and neurodevelopmental, -degenerative, and -psychiatric conditions.
Collapse
Affiliation(s)
- Laura M J Fernandez
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
A novel phospho-modulatory mechanism contributes to the calcium-dependent regulation of T-type Ca 2+ channels. Sci Rep 2019; 9:15642. [PMID: 31666636 PMCID: PMC6821770 DOI: 10.1038/s41598-019-52194-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 10/13/2019] [Indexed: 11/08/2022] Open
Abstract
Cav3 / T-type Ca2+ channels are dynamically regulated by intracellular Ca2+ ions, which inhibit Cav3 availability. Here, we demonstrate that this inhibition becomes irreversible in the presence of non-hydrolysable ATP analogs, resulting in a strong hyperpolarizing shift in the steady-state inactivation of the residual Cav3 current. Importantly, the effect of these ATP analogs was prevented in the presence of intracellular BAPTA. Additional findings obtained using intracellular dialysis of inorganic phosphate and alkaline phosphatase or NaN3 treatment further support the involvement of a phosphorylation mechanism. Contrasting with Cav1 and Cav2 Ca2+ channels, the Ca2+-dependent modulation of Cav3 channels appears to be independent of calmodulin, calcineurin and endocytic pathways. Similar findings were obtained for the native T-type Ca2+ current recorded in rat thalamic neurons of the central medial nucleus. Overall, our data reveal a new Ca2+ sensitive phosphorylation-dependent mechanism regulating Cav3 channels, with potentially important physiological implications for the multiple cell functions controlled by T-type Ca2+ channels.
Collapse
|
31
|
Mäki-Marttunen T, Kaufmann T, Elvsåshagen T, Devor A, Djurovic S, Westlye LT, Linne ML, Rietschel M, Schubert D, Borgwardt S, Efrim-Budisteanu M, Bettella F, Halnes G, Hagen E, Næss S, Ness TV, Moberget T, Metzner C, Edwards AG, Fyhn M, Dale AM, Einevoll GT, Andreassen OA. Biophysical Psychiatry-How Computational Neuroscience Can Help to Understand the Complex Mechanisms of Mental Disorders. Front Psychiatry 2019; 10:534. [PMID: 31440172 PMCID: PMC6691488 DOI: 10.3389/fpsyt.2019.00534] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is the most complex of human organs, and the pathophysiology underlying abnormal brain function in psychiatric disorders is largely unknown. Despite the rapid development of diagnostic tools and treatments in most areas of medicine, our understanding of mental disorders and their treatment has made limited progress during the last decades. While recent advances in genetics and neuroscience have a large potential, the complexity and multidimensionality of the brain processes hinder the discovery of disease mechanisms that would link genetic findings to clinical symptoms and behavior. This applies also to schizophrenia, for which genome-wide association studies have identified a large number of genetic risk loci, spanning hundreds of genes with diverse functionalities. Importantly, the multitude of the associated variants and their prevalence in the healthy population limit the potential of a reductionist functional genetics approach as a stand-alone solution to discover the disease pathology. In this review, we outline the key concepts of a "biophysical psychiatry," an approach that employs large-scale mechanistic, biophysics-founded computational modelling to increase transdisciplinary understanding of the pathophysiology and strive toward robust predictions. We discuss recent scientific advances that allow a synthesis of previously disparate fields of psychiatry, neurophysiology, functional genomics, and computational modelling to tackle open questions regarding the pathophysiology of heritable mental disorders. We argue that the complexity of the increasing amount of genetic data exceeds the capabilities of classical experimental assays and requires computational approaches. Biophysical psychiatry, based on modelling diseased brain networks using existing and future knowledge of basic genetic, biochemical, and functional properties on a single neuron to a microcircuit level, may allow a leap forward in deriving interpretable biomarkers and move the field toward novel treatment options.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Anna Devor
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dirk Schubert
- Cognitive Neuroscience Department, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Stefan Borgwardt
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Magdalena Efrim-Budisteanu
- Prof. Dr. Alex. Obregia Clinical Hospital of Psychiatry, Bucharest, Romania
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Francesco Bettella
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Geir Halnes
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Espen Hagen
- Department of Physics, University of Oslo, Oslo, Norway
| | - Solveig Næss
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Torbjørn V. Ness
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir Moberget
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christoph Metzner
- Centre for Computer Science and Informatics Research, University of Hertfordshire, Hatfield, United Kingdom
- Institute of Software Engineering and Theoretical Computer Science, Technische Universität zu Berlin, Berlin, Germany
| | - Andrew G. Edwards
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Anders M. Dale
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Gaute T. Einevoll
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Andrade A, Brennecke A, Mallat S, Brown J, Gomez-Rivadeneira J, Czepiel N, Londrigan L. Genetic Associations between Voltage-Gated Calcium Channels and Psychiatric Disorders. Int J Mol Sci 2019; 20:E3537. [PMID: 31331039 PMCID: PMC6679227 DOI: 10.3390/ijms20143537] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022] Open
Abstract
Psychiatric disorders are mental, behavioral or emotional disorders. These conditions are prevalent, one in four adults suffer from any type of psychiatric disorders world-wide. It has always been observed that psychiatric disorders have a genetic component, however, new methods to sequence full genomes of large cohorts have identified with high precision genetic risk loci for these conditions. Psychiatric disorders include, but are not limited to, bipolar disorder, schizophrenia, autism spectrum disorder, anxiety disorders, major depressive disorder, and attention-deficit and hyperactivity disorder. Several risk loci for psychiatric disorders fall within genes that encode for voltage-gated calcium channels (CaVs). Calcium entering through CaVs is crucial for multiple neuronal processes. In this review, we will summarize recent findings that link CaVs and their auxiliary subunits to psychiatric disorders. First, we will provide a general overview of CaVs structure, classification, function, expression and pharmacology. Next, we will summarize tools to study risk loci associated with psychiatric disorders. We will examine functional studies of risk variations in CaV genes when available. Finally, we will review pharmacological evidence of the use of CaV modulators to treat psychiatric disorders. Our review will be of interest for those studying pathophysiological aspects of CaVs.
Collapse
Affiliation(s)
- Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA.
| | - Ashton Brennecke
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Shayna Mallat
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Julian Brown
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | | | - Natalie Czepiel
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Laura Londrigan
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
33
|
Increased Thalamocortical Connectivity in Schizophrenia Correlates With Sleep Spindle Deficits: Evidence for a Common Pathophysiology. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2019; 4:706-714. [PMID: 31262708 DOI: 10.1016/j.bpsc.2019.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/06/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Converging evidence implicates abnormal thalamocortical interactions in the pathophysiology of schizophrenia. This evidence includes consistent findings of increased resting-state functional connectivity of the thalamus with somatosensory and motor cortex during wake and reduced spindle activity during sleep. We hypothesized that these abnormalities would be correlated, reflecting a common mechanism: reduced inhibition of thalamocortical neurons by the thalamic reticular nucleus (TRN). The TRN is the major inhibitory nucleus of the thalamus and is abnormal in schizophrenia. Reduced TRN inhibition would be expected to lead to increased and less filtered thalamic relay of sensory and motor information to the cortex during wake and reduced burst firing necessary for spindle initiation during sleep. METHODS Overnight polysomnography and resting-state functional connectivity magnetic resonance imaging were performed in 26 outpatients with schizophrenia and 30 demographically matched healthy individuals. We examined the relations of sleep spindle density during stage 2 non-rapid eye movement sleep with connectivity of the thalamus to the cortex during wakeful rest. RESULTS As in prior studies, patients with schizophrenia exhibited increased functional connectivity of the thalamus with bilateral somatosensory and motor cortex and reduced sleep spindle density. Spindle density inversely correlated with thalamocortical connectivity, including in somotosensory and motor cortex, regardless of diagnosis. CONCLUSIONS These findings link two biomarkers of schizophrenia-the sleep spindle density deficit and abnormally increased thalamocortical functional connectivity-and point to deficient TRN inhibition as a plausible mechanism. If TRN-mediated thalamocortical dysfunction increases risk for schizophrenia and contributes to its manifestations, understanding its mechanism could guide the development of targeted interventions.
Collapse
|
34
|
Abstract
De Novo Pathogenic Variants in CACNA1E Cause Developmental and Epileptic Encephalopathy With Contractures, Macrocephaly, and Dyskinesias Helbig KL, Lauerer RJ, Bahr JC, et al. Am J Hum Genet. 2019;104(3):562. Developmental and epileptic encephalopathies (DEEs) are severe neurodevelopmental disorders often beginning in infancy or early childhood that are characterized by intractable seizures, abundant epileptiform activity on electroencephalogram (EEG), and developmental impairment or regression. CACNA1E is highly expressed in the central nervous system and encodes the α1-subunit of the voltage-gated CaV2.3 channel, which conducts high-voltage-activated R-type calcium currents that initiate synaptic transmission. Using next-generation sequencing techniques, we identified de novo CACNA1E variants in 30 individuals with DEE, characterized by refractory infantile-onset seizures, severe hypotonia, and profound developmental impairment, often with congenital contractures, macrocephaly, hyperkinetic movement disorders, and early death. Most of the 14, partially recurring, variants cluster within the cytoplasmic ends of all 4 S6 segments, which form the presumed CaV2.3 channel activation gate. Functional analysis of several S6 variants revealed consistent gain-of-function effects comprising facilitated voltage-dependent activation and slowed inactivation. Another variant located in the domain II S4-S5 linker results in facilitated activation and increased current density. Five participants achieved seizure freedom on the antiepileptic drug topiramate, which blocks R-type calcium channels. We establish pathogenic variants in CACNA1E as a cause of DEEs and suggest facilitated R-type calcium currents as a disease mechanism for human epilepsy and developmental disorders.
Collapse
|
35
|
Thankachan S, Katsuki F, McKenna JT, Yang C, Shukla C, Deisseroth K, Uygun DS, Strecker RE, Brown RE, McNally JM, Basheer R. Thalamic Reticular Nucleus Parvalbumin Neurons Regulate Sleep Spindles and Electrophysiological Aspects of Schizophrenia in Mice. Sci Rep 2019; 9:3607. [PMID: 30837664 PMCID: PMC6401113 DOI: 10.1038/s41598-019-40398-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 02/11/2019] [Indexed: 02/05/2023] Open
Abstract
The thalamic reticular nucleus (TRN) is implicated in schizophrenia pathology. However, it remains unclear whether alterations of TRN activity can account for abnormal electroencephalographic activity observed in patients, namely reduced spindles (10-15 Hz) during sleep and increased delta (0.5-4 Hz) and gamma-band activity (30-80 Hz) during wakefulness. Here, we utilized optogenetic and reverse-microdialysis approaches to modulate activity of the major subpopulation of TRN GABAergic neurons, which express the calcium-binding protein parvalbumin (PV), and are implicated in schizophrenia dysfunction. An automated algorithm with enhanced efficiency and reproducibility compared to manual detection was used for sleep spindle assessment. A novel, low power, waxing-and-waning optogenetic stimulation paradigm preferentially induced spindles that were indistinguishable from spontaneously occurring sleep spindles without altering the behavioral state, when compared to a single pulse laser stimulation used by us and others. Direct optogenetic inhibition of TRN-PV neurons was ineffective in blocking spindles but increased both wakefulness and cortical delta/gamma activity, as well as impaired the 40 Hz auditory steady-state response. For the first time we demonstrate that spindle density is markedly reduced by (i) optogenetic stimulation of a major GABA/PV inhibitory input to TRN arising from basal forebrain parvalbumin neurons (BF-PV) and; (ii) localized pharmacological inhibition of low-threshold calcium channels, implicated as a genetic risk factor for schizophrenia. Together with clinical findings, our results support impaired TRN-PV neuron activity as a potential cause of schizophrenia-linked abnormalities in cortical delta, gamma, and spindle activity. Modulation of the BF-PV input to TRN may improve these neural abnormalities.
Collapse
Affiliation(s)
- Stephen Thankachan
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Fumi Katsuki
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - James T McKenna
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Chun Yang
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Charu Shukla
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Karl Deisseroth
- Stanford University, Psychiatry and Behavioral Sciences/Bioengineering, Stanford, CA, USA
| | - David S Uygun
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Robert E Strecker
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - Ritchie E Brown
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA
| | - James M McNally
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA.
| | - Radhika Basheer
- VA Boston Healthcare System and Harvard Medical School, Dept. of Psychiatry, West Roxbury, MA, USA.
| |
Collapse
|
36
|
T-type calcium channels: From molecule to therapeutic opportunities. Int J Biochem Cell Biol 2019; 108:34-39. [DOI: 10.1016/j.biocel.2019.01.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
|
37
|
Abstract
There is overwhelming evidence that sleep is crucial for memory consolidation. Patients with schizophrenia and their unaffected relatives have a specific deficit in sleep spindles, a defining oscillation of non-rapid eye movement (NREM) Stage 2 sleep that, in coordination with other NREM oscillations, mediate memory consolidation. In schizophrenia, the spindle deficit correlates with impaired sleep-dependent memory consolidation, positive symptoms, and abnormal thalamocortical connectivity. These relations point to dysfunction of the thalamic reticular nucleus (TRN), which generates spindles, gates the relay of sensory information to the cortex, and modulates thalamocortical communication. Genetic studies are beginning to provide clues to possible neurodevelopmental origins of TRN-mediated thalamocortical circuit dysfunction and to identify novel targets for treating the related memory deficits and symptoms. By forging empirical links in causal chains from risk genes to thalamocortical circuit dysfunction, spindle deficits, memory impairment, symptoms, and diagnosis, future research can advance our mechanistic understanding, treatment, and prevention of schizophrenia.
Collapse
Affiliation(s)
- Dara S Manoach
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; .,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Robert Stickgold
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215;
| |
Collapse
|
38
|
Mäki-Marttunen T, Krull F, Bettella F, Hagen E, Næss S, Ness TV, Moberget T, Elvsåshagen T, Metzner C, Devor A, Edwards AG, Fyhn M, Djurovic S, Dale AM, Andreassen OA, Einevoll GT. Alterations in Schizophrenia-Associated Genes Can Lead to Increased Power in Delta Oscillations. Cereb Cortex 2019; 29:875-891. [PMID: 30475994 PMCID: PMC6319172 DOI: 10.1093/cercor/bhy291] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies have implicated many ion channels in schizophrenia pathophysiology. Although the functions of these channels are relatively well characterized by single-cell studies, the contributions of common variation in these channels to neurophysiological biomarkers and symptoms of schizophrenia remain elusive. Here, using computational modeling, we show that a common biomarker of schizophrenia, namely, an increase in delta-oscillation power, may be a direct consequence of altered expression or kinetics of voltage-gated ion channels or calcium transporters. Our model of a circuit of layer V pyramidal cells highlights multiple types of schizophrenia-related variants that contribute to altered dynamics in the delta-frequency band. Moreover, our model predicts that the same membrane mechanisms that increase the layer V pyramidal cell network gain and response to delta-frequency oscillations may also cause a deficit in a single-cell correlate of the prepulse inhibition, which is a behavioral biomarker highly associated with schizophrenia.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Simula Research Laboratory, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Florian Krull
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Francesco Bettella
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Hagen
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Solveig Næss
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Torbjørn V Ness
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Torgeir Moberget
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Christoph Metzner
- Centre for Computer Science and Informatics Research, University of Hertfordshire, Hatfield, UK
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Gaute T Einevoll
- Department of Physics, University of Oslo, Oslo, Norway
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
39
|
Bygrave AM, Kilonzo K, Kullmann DM, Bannerman DM, Kätzel D. Can N-Methyl-D-Aspartate Receptor Hypofunction in Schizophrenia Be Localized to an Individual Cell Type? Front Psychiatry 2019; 10:835. [PMID: 31824347 PMCID: PMC6881463 DOI: 10.3389/fpsyt.2019.00835] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/21/2019] [Indexed: 01/07/2023] Open
Abstract
Hypofunction of N-methyl-D-aspartate glutamate receptors (NMDARs), whether caused by endogenous factors like auto-antibodies or mutations, or by pharmacological or genetic manipulations, produces a wide variety of deficits which overlap with-but do not precisely match-the symptom spectrum of schizophrenia. In order to understand how NMDAR hypofunction leads to different components of the syndrome, it is necessary to take into account which neuronal subtypes are particularly affected by it in terms of detrimental functional alterations. We provide a comprehensive overview detailing findings in rodent models with cell type-specific knockout of NMDARs. Regarding inhibitory cortical cells, an emerging model suggests that NMDAR hypofunction in parvalbumin (PV) positive interneurons is a potential risk factor for this disease. PV interneurons display a selective vulnerability resulting from a combination of genetic, cellular, and environmental factors that produce pathological multi-level positive feedback loops. Central to this are two antioxidant mechanisms-NMDAR activity and perineuronal nets-which are themselves impaired by oxidative stress, amplifying disinhibition. However, NMDAR hypofunction in excitatory pyramidal cells also produces a range of schizophrenia-related deficits, in particular maladaptive learning and memory recall. Furthermore, NMDAR blockade in the thalamus disturbs thalamocortical communication, and NMDAR ablation in dopaminergic neurons may provoke over-generalization in associative learning, which could relate to the positive symptom domain. Therefore, NMDAR hypofunction can produce schizophrenia-related effects through an action on various different circuits and cell types.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Kasyoka Kilonzo
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| | - Dimitri M Kullmann
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Dennis Kätzel
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
40
|
Zhang T, Zhu L, Ni T, Liu D, Chen G, Yan Z, Lin H, Guan F, Rice JP. Voltage-gated calcium channel activity and complex related genes and schizophrenia: A systematic investigation based on Han Chinese population. J Psychiatr Res 2018; 106:99-105. [PMID: 30308413 DOI: 10.1016/j.jpsychires.2018.09.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/19/2022]
Abstract
Schizophrenia (SCZ) is a devastating mental disorder affecting approximately 1% of the worldwide population. Early studies have indicated that genetics plays an important role in the onset and development of SCZ. Accumulating evidence supports that SCZ is linked to abnormalities of synapse transmission and synaptic plasticity. Voltage-gated calcium channel (VGCC) subunits are critical for mediating intracellular Ca2 + influx and therefore are responsible for changing neuronal excitability and synaptic plasticity. To systematically investigate the role of calcium signaling genes in SCZ susceptibility, we conducted a case-control study that included 2518 SCZ patients and 7521 healthy controls with Chinese Han ancestry. Thirty-seven VGCC genes, including 363 tag single nucleotide polymorphisms (SNPs), were examined. Our study replicated the following previously identified susceptible loci: CACNA1C, CACNB2, OPRM1, GRM7 and PDE4B. In addition, several novel loci including CACNA2D1, PDE4D, NALCN, and CACNA2D3 were also identified to be associated with SCZ in our Han Chinese sample. Combined with GTEx eQTL data, we have shown that CASQ2, ITGAV, and TMC2 can be also added into the prioritization list of SCZ susceptible genes. Two-way interaction analyses identified widespread gene-by-gene interactions among VGCC activity and complex-related genes for the susceptibility of SCZ. Further sequencing based studies are still needed to unravel potential contributions of schizophrenia risk from rare or low frequency variants of these candidate genes.
Collapse
Affiliation(s)
- Tianxiao Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Li Zhu
- Department of Forensic Psychiatry, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Tong Ni
- Department of Forensic Psychiatry, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Dan Liu
- Department of Forensic Psychiatry, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Gang Chen
- Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Department of Forensic Pathology, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Zhilan Yan
- Department of Forensic Psychiatry, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Huali Lin
- Xi'an Mental Health Center, 15 Yanyin Road, Xi'an, Shaanxi, 710086, China
| | - Fanglin Guan
- Department of Forensic Psychiatry, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China; Key Laboratory of National Ministry of Health for Forensic Sciences, School of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| | - John P Rice
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, 63124, USA
| |
Collapse
|
41
|
Steullet P, Cabungcal JH, Bukhari SA, Ardelt MI, Pantazopoulos H, Hamati F, Salt TE, Cuenod M, Do KQ, Berretta S. The thalamic reticular nucleus in schizophrenia and bipolar disorder: role of parvalbumin-expressing neuron networks and oxidative stress. Mol Psychiatry 2018; 23:2057-2065. [PMID: 29180672 PMCID: PMC5972042 DOI: 10.1038/mp.2017.230] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 08/29/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023]
Abstract
Growing evidence points to a disruption of cortico-thalamo-cortical circuits in schizophrenia (SZ) and bipolar disorder (BD). Clues for a specific involvement of the thalamic reticular nucleus (TRN) come from its unique neuronal characteristics and neural connectivity, allowing it to shape the thalamo-cortical information flow. A direct involvement of the TRN in SZ and BD has not been tested thus far. We used a combination of human postmortem and rodent studies to test the hypothesis that neurons expressing parvalbumin (PV neurons), a main TRN neuronal population, and associated Wisteria floribunda agglutinin-labeled perineuronal nets (WFA/PNNs) are altered in SZ and BD, and that these changes may occur early in the course of the disease as a consequence of oxidative stress. In both disease groups, marked decreases of PV neurons (immunoreactive for PV) and WFA/PNNs were observed in the TRN, with no effects of duration of illness or age at onset. Similarly, in transgenic mice with redox dysregulation, numbers of PV neurons and WFA/PNN+PV neurons were decreased in transgenic compared with wild-type mice; these changes were present at postnatal day (P) 20 for PV neurons and P40 for WFA/PNN+PV neurons, accompanied by alterations of their firing properties. These results show profound abnormalities of PV neurons in the TRN of subjects with SZ and BD, and offer support for the hypothesis that oxidative stress may play a key role in impacting TRN PV neurons at early stages of these disorders. We put forth that these TRN abnormalities may contribute to disruptions of sleep spindles, focused attention and emotion processing in these disorders.
Collapse
Affiliation(s)
- Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Jan-Harry Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Syed A. Bukhari
- Translational Neuroscience Laboratory, Mclean Hospital, Belmont, MA, USA
| | | | - Harry Pantazopoulos
- Translational Neuroscience Laboratory, Mclean Hospital, Belmont, MA, USA,Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Fadi Hamati
- Translational Neuroscience Laboratory, Mclean Hospital, Belmont, MA, USA
| | - Thomas E. Salt
- The Institute of Ophthalmology, University College London, London, UK
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Kim Q. Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Sabina Berretta
- Translational Neuroscience Laboratory, Mclean Hospital, Belmont, MA, USA,Department of Psychiatry, Harvard Medical School, Boston, MA, USA,Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Tylee DS, Sun J, Hess JL, Tahir MA, Sharma E, Malik R, Worrall BB, Levine AJ, Martinson JJ, Nejentsev S, Speed D, Fischer A, Mick E, Walker BR, Crawford A, Grant SF, Polychronakos C, Bradfield JP, Sleiman PMA, Hakonarson H, Ellinghaus E, Elder JT, Tsoi LC, Trembath RC, Barker JN, Franke A, Dehghan A, The 23andMe Research Team, The Inflammation Working Group of the CHARGE Consortium, The METASTROKE Consortium of the International Stroke Genetics Consortium, The Netherlands Twin Registry, The neuroCHARGE Working Group, The Eating Disorder Working Groups of the Psychiatric Genomics Consortium, The Obsessive Compulsive Disorder and Tourette Syndrome Working Group, Faraone SV, Glatt. SJ. Genetic correlations among psychiatric and immune-related phenotypes based on genome-wide association data. Am J Med Genet B Neuropsychiatr Genet 2018; 177:641-657. [PMID: 30325587 PMCID: PMC6230304 DOI: 10.1002/ajmg.b.32652] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/21/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022]
Abstract
Individuals with psychiatric disorders have elevated rates of autoimmune comorbidity and altered immune signaling. It is unclear whether these altered immunological states have a shared genetic basis with those psychiatric disorders. The present study sought to use existing summary-level data from previous genome-wide association studies to determine if commonly varying single nucleotide polymorphisms are shared between psychiatric and immune-related phenotypes. We estimated heritability and examined pair-wise genetic correlations using the linkage disequilibrium score regression (LDSC) and heritability estimation from summary statistics methods. Using LDSC, we observed significant genetic correlations between immune-related disorders and several psychiatric disorders, including anorexia nervosa, attention deficit-hyperactivity disorder, bipolar disorder, major depression, obsessive compulsive disorder, schizophrenia, smoking behavior, and Tourette syndrome. Loci significantly mediating genetic correlations were identified for schizophrenia when analytically paired with Crohn's disease, primary biliary cirrhosis, systemic lupus erythematosus, and ulcerative colitis. We report significantly correlated loci and highlight those containing genome-wide associations and candidate genes for respective disorders. We also used the LDSC method to characterize genetic correlations among the immune-related phenotypes. We discuss our findings in the context of relevant genetic and epidemiological literature, as well as the limitations and caveats of the study.
Collapse
Affiliation(s)
- Daniel S. Tylee
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Jiayin Sun
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Jonathan L. Hess
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Muhammad A. Tahir
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Esha Sharma
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Bradford B. Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Andrew J. Levine
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, U.S.A
| | - Jeremy J. Martinson
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, PA, U.S.A
| | | | - Doug Speed
- Aarhus Institute for Advanced Studies and University College London, London, U.K
| | - Annegret Fischer
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Eric Mick
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, U.S.A
| | - Brian R. Walker
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, U.K
| | - Andrew Crawford
- School of Social and Community Medicine, MRC Integrated Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Center for Applied Genomics, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
| | - Struan F.A. Grant
- Center for Applied Genomics, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Constantin Polychronakos
- Endocrine Genetics Laboratory, Department of Pediatrics and the Child Health Program of the Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jonathan P. Bradfield
- Center for Applied Genomics, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Quantinuum Research LLC, San Diego, CA, U.S.A
| | - Patrick M. A. Sleiman
- Center for Applied Genomics, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, U.S.A
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, U.S.A
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - James T. Elder
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lam C. Tsoi
- Department of Dermatology, Veterans Affairs Hospital, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Richard C. Trembath
- Division of Genetics and Molecular Medicine, King’s College London, London, UK
| | - Jonathan N. Barker
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Abbas Dehghan
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London
| | | | | | - Stephen V. Faraone
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - Stephen J. Glatt.
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab); Departments of Psychiatry and Behavioral Sciences & Neuroscience and Physiology; SUNY Upstate Medical University; Syracuse, NY, U.S.A
| |
Collapse
|
43
|
Song JHT, Lowe CB, Kingsley DM. Characterization of a Human-Specific Tandem Repeat Associated with Bipolar Disorder and Schizophrenia. Am J Hum Genet 2018; 103:421-430. [PMID: 30100087 PMCID: PMC6128321 DOI: 10.1016/j.ajhg.2018.07.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022] Open
Abstract
Bipolar disorder (BD) and schizophrenia (SCZ) are highly heritable diseases that affect more than 3% of individuals worldwide. Genome-wide association studies have strongly and repeatedly linked risk for both of these neuropsychiatric diseases to a 100 kb interval in the third intron of the human calcium channel gene CACNA1C. However, the causative mutation is not yet known. We have identified a human-specific tandem repeat in this region that is composed of 30 bp units, often repeated hundreds of times. This large tandem repeat is unstable using standard polymerase chain reaction and bacterial cloning techniques, which may have resulted in its incorrect size in the human reference genome. The large 30-mer repeat region is polymorphic in both size and sequence in human populations. Particular sequence variants of the 30-mer are associated with risk status at several flanking single-nucleotide polymorphisms in the third intron of CACNA1C that have previously been linked to BD and SCZ. The tandem repeat arrays function as enhancers that increase reporter gene expression in a human neural progenitor cell line. Different human arrays vary in the magnitude of enhancer activity, and the 30-mer arrays associated with increased psychiatric disease risk status have decreased enhancer activity. Changes in the structure and sequence of these arrays likely contribute to changes in CACNA1C function during human evolution and may modulate neuropsychiatric disease risk in modern human populations.
Collapse
Affiliation(s)
- Janet H T Song
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Craig B Lowe
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David M Kingsley
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Marks WN, Parker ME, Zabder NK, Greba Q, Snutch TP, Howland JG. T-type calcium channels in the orbitofrontal cortex mediate sensory integration as measured using a spontaneous oddity task in rats. ACTA ACUST UNITED AC 2018; 25:317-324. [PMID: 29907639 PMCID: PMC6004062 DOI: 10.1101/lm.047332.118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/02/2018] [Indexed: 11/24/2022]
Abstract
The roles of low-voltage-activated (T-type) calcium channels in brain diseases have been studied extensively. Less is known regarding the involvement of T-type channels in cognition and behavior. Sensory integration (SI) is a cognitive process whereby the brain uses unimodal or multimodal sensory features to create a comprehensive representation of the environment. The multisensory object oddity (MSO) task assesses SI using combinations of sensory features of objects, either in the same or different sensory modalities. The regulation of SI involves the orbitofrontal cortex (OFC), an area which shows high levels of T-type calcium channel expression. We tested the effects of blocking T-type calcium channels on the MSO task with the selective T-type antagonist, Z944 (5 mg/kg; i.p. systemic; 100 or 500 µM OFC infusion), in male Long Evans rats. With systemic treatment, Z944 impaired the visual and visual-olfactory versions of the task. Infusion of 100 and 500 µM Z944 produced deficits in the olfactory version of the task. In addition, only vehicle-infused, but not Z944-infused, rats showed significant performance above chance for all task variants. Thus, the present results suggest that T-type calcium channels in OFC are involved in SI of features in an oddity task. Given that unimodal SI was disrupted by OFC infusions of Z944, the deficits in the multimodal task must be interpreted with caution. As SI is disrupted in psychiatric disorders, further investigations elucidating the brain regions implicated in SI regulation by T-type calcium channels may help inform therapeutic development for those suffering from SI impairments.
Collapse
Affiliation(s)
- Wendie N Marks
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Madeline E Parker
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Nadine K Zabder
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Quentin Greba
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - John G Howland
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
45
|
Krol A, Wimmer RD, Halassa MM, Feng G. Thalamic Reticular Dysfunction as a Circuit Endophenotype in Neurodevelopmental Disorders. Neuron 2018; 98:282-295. [PMID: 29673480 PMCID: PMC6886707 DOI: 10.1016/j.neuron.2018.03.021] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 01/30/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
Diagnoses of behavioral disorders such as autism spectrum disorder and schizophrenia are based on symptomatic descriptions that have been difficult to connect to mechanism. Although psychiatric genetics provide insight into the genetic underpinning of such disorders, with a majority of cases explained by polygenic factors, it remains difficult to design rational treatments. In this review, we highlight the value of understanding neural circuit function both as an intermediate level of explanatory description that links gene to behavior and as a pathway for developing rational diagnostics and therapeutics for behavioral disorders. As neural circuits perform hierarchically organized computational functions and give rise to network-level processes (e.g., macroscopic rhythms and goal-directed or homeostatic behaviors), correlated network-level deficits may indicate perturbation of a specific circuit. Therefore, identifying such correlated deficits or a circuit endophenotype would provide a mechanistic point of entry, enhancing both diagnosis and treatment of a given behavioral disorder. We focus on a circuit endophenotype of the thalamic reticular nucleus (TRN) and how its impairment in neurodevelopmental disorders gives rise to a correlated set of readouts across sleep and attention. Because TRN neurons express several disorder-relevant genes identified through genome-wide association studies, exploring the consequences of different TRN disruptions may be of broad translational significance.
Collapse
Affiliation(s)
- Alexandra Krol
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ralf D Wimmer
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael M Halassa
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
46
|
DeMichele-Sweet MAA, Weamer EA, Klei L, Vrana DT, Hollingshead DJ, Seltman HJ, Sims R, Foroud T, Hernandez I, Moreno-Grau S, Tárraga L, Boada M, Ruiz A, Williams J, Mayeux R, Lopez OL, Sibille EL, Kamboh MI, Devlin B, Sweet RA. Genetic risk for schizophrenia and psychosis in Alzheimer disease. Mol Psychiatry 2018; 23:963-972. [PMID: 28461698 PMCID: PMC5668212 DOI: 10.1038/mp.2017.81] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 12/29/2022]
Abstract
Psychotic symptoms, defined as the occurrence of delusions or hallucinations, are frequent in Alzheimer disease (AD), affecting ~40 to 60% of individuals with AD (AD with psychosis (AD+P)). In comparison with AD subjects without psychosis, AD+P subjects have more rapid cognitive decline and poor outcomes. Prior studies have estimated the heritability of psychosis in AD at 61%, but the underlying genetic sources of this risk are not known. We evaluated a Discovery Cohort of 2876 AD subjects with (N=1761) or without psychosis (N=1115). All subjects were genotyped using a custom genotyping array designed to evaluate single-nucleotide polymorphisms (SNPs) with evidence of genetic association with AD+P and include SNPs affecting or putatively affecting risk for schizophrenia and AD. Results were replicated in an independent cohort of 2194 AD subjects with (N=734) or without psychosis (N=1460). We found that AD+P is associated with polygenic risk for a set of novel loci and inversely associated with polygenic risk for schizophrenia. Among the biologic pathways identified by the associations of schizophrenia SNPs with AD+P are endosomal trafficking, autophagy and calcium channel signaling. To the best of our knowledge, these findings provide the first clear demonstration that AD+P is associated with common genetic variation. In addition, they provide an unbiased link between polygenic risk for schizophrenia and a lower risk of psychosis in AD. This provides an opportunity to leverage progress made in identifying the biologic effects of schizophrenia alleles to identify novel mechanisms protecting against more rapid cognitive decline and psychosis risk in AD.
Collapse
Affiliation(s)
| | - Elise A. Weamer
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| | - Lambertus Klei
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Dylan T. Vrana
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA
| | - Deborah J. Hollingshead
- Genomics Research Core of the Health Sciences Core Research Facilities, University of Pittsburgh, Pittsburgh, PA
| | - Howard J. Seltman
- Department of Statistics, Carnegie Mellon University, Pittsburgh, PA
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - Tatiana Foroud
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Isabel Hernandez
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Sonia Moreno-Grau
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Lluís Tárraga
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Mercè Boada
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Agustin Ruiz
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Julie Williams
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - Richard Mayeux
- Departments of Neurology, Psychiatry and Epidemiology, Columbia University, New York, NY
| | - Oscar L. Lopez
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| | - Etienne L. Sibille
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Departments of Psychiatry and of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
- VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC) VA Pittsburgh Healthcare System, Pittsburgh, PA
| |
Collapse
|
47
|
Scolnick EM. The Path to New Therapies for Schizophrenia and Bipolar Illness. FASEB J 2017; 31:1254-1259. [PMID: 28360375 DOI: 10.1096/fj.201700028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/19/2017] [Indexed: 11/11/2022]
Abstract
Schizophrenia and bipolar illness are two of the most serious forms of mental illness. Until relatively recently, almost nothing was known about the molecular pathogenesis of either illness. The single largest risk factor that predisposes people to schizophrenia or bipolar illness is genetic risk. Heritability is high, and the incidence is significantly higher in identical twins than in nonidentical twins. Despite decades of work aimed at identifying the genes involved in these two illnesses, virtually no progress had been made until the past decade. With the knowledge and technologies that have been gained from the Human Genome Project, it has been possible to begin to understand the underlying genetics and to use the new information to begin the effort to discover new and better medicines to treat these illnesses. This article will describe the past decade of work toward this goal and articulate both the promise that now exists and what is still needed to bring dramatic and tangible change to patients.-Scolnick, E. M. The path to new therapies for schizophrenia and bipolar illness.
Collapse
Affiliation(s)
- Edward M Scolnick
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
48
|
Xu W, Liu Y, Chen J, Guo Q, Liu K, Wen Z, Zhou Z, Song Z, Zhou J, He L, Yi Q, Shi Y. Genetic risk between the CACNA1I gene and schizophrenia in Chinese Uygur population. Hereditas 2017; 155:5. [PMID: 28725167 PMCID: PMC5513035 DOI: 10.1186/s41065-017-0037-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/07/2017] [Indexed: 01/28/2023] Open
Abstract
Background Schizophrenia (SCZ) is a common mental disorder with high heritability, and genetic factors play a major role in the pathogenesis. Recent researches indicated that the CACNA1I involved in calcium channels probably affect the potential pathogenesis of SCZ. Results In this study, we attempted to investigate whether the CACNA1I gene contributes the risk to SCZ in the Uighur Chinese population, and performed a case-control study involving 985 patient samples and 1218 normal controls to analyze nine SNPs within the CACNA1I gene. Among these sites, six SNPs were significantly associated with SCZ in the allele distribution: rs132575 (adjusted Pallele = 0.039, OR = 1.159), rs713860 (adjusted Pallele = 0.039, OR = 0.792), rs738168 (adjusted Pallele = 0.039, OR = 0.785), rs136805 (adjusted Pallele = 0.014, OR = 1.212), rs5757760 (adjusted Pallele = 0.042, OR = 0.873) and rs5750871 (adjusted Pallele = 0.039, OR = 0.859). In addition, two SNPs turned to be risk factors for SCZ not only in the allele distribution, but also in the genotype distribution: rs132575 (adjusted Pgenotype = 0.037) and rs136805 (adjusted Pgenotype = 0.037). Conclusions Overall, the present study provided evidence that significant association exists between the CACNA1I gene and SCZ in the Uighur Chinese population, subsequent validation of functional analysis and genetic association studies are needed to further extend this study.
Collapse
Affiliation(s)
- Wei Xu
- Department of biology, School of Life Science, Anhui Medical University, 81 meishan road, Hefei, Anhui 230031 China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Yahui Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030 People's Republic of China
| | - Qingli Guo
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Ke Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Zujia Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Zhaowei Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Zhijian Song
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Lin He
- Department of biology, School of Life Science, Anhui Medical University, 81 meishan road, Hefei, Anhui 230031 China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| | - Qizhong Yi
- Psychological Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Xinjiang, 830054 China
| | - Yongyong Shi
- Department of biology, School of Life Science, Anhui Medical University, 81 meishan road, Hefei, Anhui 230031 China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, 200030 People's Republic of China
| |
Collapse
|
49
|
Lazniewska J, Weiss N. Glycosylation of voltage-gated calcium channels in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:662-668. [PMID: 28109749 DOI: 10.1016/j.bbamem.2017.01.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 12/26/2022]
Abstract
Voltage-gated calcium channels (VGCCs) are transmembrane proteins that translate electrical activities into intracellular calcium elevations and downstream signaling pathways. They serve essential physiological functions including communication between nerve cells, muscle contraction, cardiac activity, and release of hormones and neurotransmitters. Asparagine-linked glycosylation has emerged as an essential post-translational modification to control the number of channels embedded in the plasma membrane but also their functional gating properties. This review provides a comprehensive overview about the current state of knowledge on the role of glycosylation in the expression and functioning of VGCCs, and discusses how variations in the glycosylation of the channel proteins can contribute to pathological conditions.
Collapse
Affiliation(s)
- Joanna Lazniewska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|