1
|
Allen O, Coombes BJ, Pazdernik V, Gisabella B, Hartley J, Biernacka JM, Frye MA, Markota M, Pantazopoulos H. Differential serum levels of CACNA1C, circadian rhythm and stress response molecules in subjects with bipolar disorder: Associations with genetic and clinical factors. J Affect Disord 2024; 367:148-156. [PMID: 39233237 PMCID: PMC11496001 DOI: 10.1016/j.jad.2024.08.238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/23/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Many patients with bipolar disorder (BD) do not respond to or have difficulties tolerating lithium and/or other mood stabilizing agents. There is a need for personalized treatments based on biomarkers in guiding treatment options. The calcium voltage-gated channel CACNA1C is a promising candidate for developing personalized treatments. CACNA1C is implicated in BD by genome-wide association studies and several lines of evidence suggest that targeting L-type calcium channels could be an effective treatment strategy. However, before such individualized treatments can be pursued, biomarkers predicting treatment response need to be developed. METHODS As a first step in testing the hypothesis that CACNA1C genotype is associated with serum levels of CACNA1C, we conducted ELISA measures on serum samples from 100 subjects with BD and 100 control subjects. RESULTS We observed significantly higher CACNA1C (p < 0.01) protein levels in subjects with BD. The risk single nucleotide polymorpshism (SNP) (rs11062170) showed functional significance as subjects homozygous for the risk allele (CC) had significantly greater CACNA1C protein levels compared to subjects with one (p = 0.013) or no copies (p = 0.009). We observed higher somatostatin (SST) (p < 0.003) protein levels and lower levels of the clock protein aryl hydrocarbon receptor nuclear translocator-like (ARTNL) (p < 0.03) and stress signaling factor corticotrophin releasing hormone (CRH) (p < 0.001) in BD. SST and period 2 (PER2) protein levels were associated with both alcohol dependence and lithium response. CONCLUSIONS Our findings represent the first evidence for increased serum levels of CACNA1C in BD. Along with altered levels of SST, ARNTL, and CRH our findings suggest CACNA1C is associated with circadian rhythm and stress response disturbances in BD.
Collapse
Affiliation(s)
- Obie Allen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Brandon J Coombes
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Vanessa Pazdernik
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA; Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Joshua Hartley
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Joanna M Biernacka
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Matej Markota
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA; Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
2
|
Kamp D. A physical perspective on lithium therapy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:55-74. [PMID: 39547449 DOI: 10.1016/j.pbiomolbio.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
Lithium salts have strong medical properties in neurological disorders such as bipolar disorder and lithium-responsive headaches. They have recently gathered attention due to their potential preventive effect in viral infections. Though the therapeutic effect of lithium was documented by Cade in the late 1940s, its underlying mechanism of action is still disputed. Acute lithium exposure has an activating effect on excitable organic tissue and organisms, and is highly toxic. Lithium exposure is associated with a strong metabolic response in the organism, with large changes in phospholipid and cholesterol expression. Opposite to acute exposure, this metabolic response alleviates excessive cellular activity. The presence of lithium ions strongly affects lipid conformation and membrane phase unlike other alkali ions, with consequences for membrane permeability, buffer property and excitability. This review investigates how lithium ions affect lipid membrane composition and function, and how lithium response might in fact be the body's attempt to counteract the physical presence of lithium ions at cell level. Ideas for further research in microbiology and drug development are discussed.
Collapse
Affiliation(s)
- Dana Kamp
- The Niels Bohr Institute, Copenhagen University, Copenhagen, Denmark.
| |
Collapse
|
3
|
Sriretnakumar V, Harripaul R, Kennedy JL, So J. When rare meets common: Treatable genetic diseases are enriched in the general psychiatric population. Am J Med Genet A 2024; 194:e63609. [PMID: 38532509 DOI: 10.1002/ajmg.a.63609] [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: 11/02/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Mental illnesses are one of the biggest contributors to the global disease burden. Despite the increased recognition, diagnosis and ongoing research of mental health disorders, the etiology and underlying molecular mechanisms of these disorders are yet to be fully elucidated. Moreover, despite many treatment options available, a large subset of the psychiatric patient population is nonresponsive to standard medications and therapies. There has not been a comprehensive study to date examining the burden and impact of treatable genetic disorders (TGDs) that can present with neuropsychiatric features in psychiatric patient populations. In this study, we test the hypothesis that TGDs that present with psychiatric symptoms are more prevalent within psychiatric patient populations compared to the general population by performing targeted next-generation sequencing of 129 genes associated with 108 TGDs in a cohort of 2301 psychiatric patients. In total, 48 putative affected and 180 putative carriers for TGDs were identified, with known or likely pathogenic variants in 79 genes. Despite screening for only 108 genetic disorders, this study showed a two-fold (2.09%) enrichment for genetic disorders within the psychiatric population relative to the estimated 1% cumulative prevalence of all single gene disorders globally. This strongly suggests that the prevalence of these, and most likely all, genetic diseases is greatly underestimated in psychiatric populations. Increasing awareness and ensuring accurate diagnosis of TGDs will open new avenues to targeted treatment for a subset of psychiatric patients.
Collapse
Affiliation(s)
- Venuja Sriretnakumar
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Ricardo Harripaul
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Joyce So
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Genetics, Departments of Medicine and Pediatrics, University of California, San Francisco, California, USA
| |
Collapse
|
4
|
Dickerson MR, Reed J. Pharmacogenetic testing may benefit people receiving low-dose lithium in clinical practice. J Am Assoc Nurse Pract 2024; 36:320-328. [PMID: 37882688 DOI: 10.1097/jxx.0000000000000968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Mental illnesses are leading causes of disability in the United States. Some evidence supports that pharmacogenetic testing may be beneficial in select populations and that lithium is beneficial for treating mood disorders and anxiety in some populations. PURPOSE This research aimed to determine whether low-dose lithium effectively decreases depression and anxiety in adults with a risk allele for CACNA1C genotypes. METHODOLOGY The study design was correlational. Fifty patients were treated at a nurse practitioner-owned clinic in Prairie Village, Kansas. Chart review was used. Adults older than 18 years diagnosed with major depressive disorder, bipolar disorder, or generalized anxiety disorder presenting with an abnormality in the CACNA1C gene single-nucleotide polymorphism rs1006737 were included in this research. Assessment tools used were the Patient Health Questionnaire-9 for depression and GAD-7 for anxiety. RESULTS Low-dose lithium significantly decreased depression by 66% ( p < .001) and anxiety by 65% ( p = <.001). There was a significant difference in pretest depression levels based on CACNA1C genotype ( p = .033). The A allele frequency was 60% higher (48%) in this population than found in general population (30%). CONCLUSIONS Low-dose lithium significantly decreased anxiety and depression compared with baseline. People with different versions of the CACNA1C genotype had responses that differed significantly. The A risk allele was 60% more common than in the general population. IMPLICATIONS This study could aid in establishing genetic testing as an effective clinical tool for treating depression and anxiety using lithium, an inexpensive and widely available medication.
Collapse
Affiliation(s)
- Michael Ray Dickerson
- University of Missouri-Kansas City, Kansas City, Missouri
- Southwest Baptist University, Springfield, Missouri
| | | |
Collapse
|
5
|
Allen O, Coombes BJ, Pazdernik V, Gisabella B, Hartley J, Biernacka JM, Frye MA, Markota M, Pantazopoulos H. Differential Serum Levels of CACNA1C, Circadian Rhythm and Stress Response Molecules in Subjects with Bipolar Disorder: Associations with Genetic and Clinical Factors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.11.24305678. [PMID: 38645236 PMCID: PMC11030295 DOI: 10.1101/2024.04.11.24305678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Many patients with bipolar disorder (BD) do not respond to or have difficulties tolerating lithium and/or other mood stabilizing agents. There is a need for personalized treatments based on biomarkers in guiding treatment options. The calcium voltage-gated channel CACNA1C is a promising candidate for developing personalized treatments. CACNA1C is implicated in BD by genome-wide association studies and several lines of evidence suggest that targeting L-type calcium channels could be an effective treatment strategy. However, before such individualized treatments can be pursued, biomarkers predicting treatment response need to be developed. Methods As a first step in testing the hypothesis that CACNA1C genotype is associated with serum levels of CACNA1C, we conducted ELISA measures on serum samples from 100 subjects with BD and 100 control subjects. Results We observed significantly higher CACNA1C (p<0.01) protein levels in subjects with BD. The risk SNP (rs11062170) showed functional significance as subjects homozygous for the risk allele (CC) had significantly greater CACNA1C protein levels compared to subjects with one (p=0.013) or no copies (p=0.009). We observed higher somatostatin (SST) (p<0.003) protein levels and lower levels of the clock protein ARTNL (p<0.03) and stress signaling factor corticotrophin releasing hormone (CRH) (p<0.001) in BD. SST and PER2 protein levels were associated with both alcohol dependence and lithium response. Conclusions Our findings represent the first evidence for increased serum levels of CACNA1C in BD. Along with altered levels of SST, ARNTL, and CRH our findings suggest CACNA1C is associated with circadian rhythm and stress response disturbances in BD.
Collapse
Affiliation(s)
- Obie Allen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | - Brandon J. Coombes
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Vanessa Pazdernik
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua Hartley
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joanna M. Biernacka
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Matej Markota
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
6
|
Bukhteeva I, Rahman FA, Kendall B, Duncan RE, Quadrilatero J, Pavlov EV, Gingras MJP, Leonenko Z. Effects of lithium isotopes on sodium/lithium co-transport and calcium efflux through the sodium/calcium/lithium exchanger in mitochondria. Front Physiol 2024; 15:1354091. [PMID: 38655027 PMCID: PMC11036541 DOI: 10.3389/fphys.2024.1354091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/06/2024] [Indexed: 04/26/2024] Open
Abstract
The effects of lithium (Li) isotopes and their impact on biological processes have recently gained increased attention due to the significance of Li as a pharmacological agent and the potential that Li isotopic effects in neuroscience contexts may constitute a new example of quantum effects in biology. Previous studies have shown that the two Li isotopes, which differ in mass and nuclear spin, have unusual different effects in vivo and in vitro and, although some molecular targets for Li isotope fractionation have been proposed, it is not known whether those result in observable downstream neurophysiological effects. In this work we studied fluxes of Li+, sodium (Na+) and calcium (Ca2+) ions in the mitochondrial sodium/calcium/lithium exchanger (NCLX), the only transporter known with recognized specificity for Li+. We studied the effect of Li+ isotopes on Ca2+ efflux from heart mitochondria in comparison to natural Li+ and Na+ using Ca2+-induced fluorescence and investigated a possible Li isotope fractionation in mitochondria using inductively coupled plasma mass spectrometry (ICP-MS). Our fluorescence data indicate that Ca2+ efflux increases with higher concentrations of either Li+ or Na+. We found that the simultaneous presence of Li+ and Na+ increases Ca2+ efflux compared to Ca2+ efflux caused by the same concentration of Li+ alone. However, no differentiation in the Ca2+ efflux between the two Li+ isotopes was observed, either for Li+ alone or in mixtures of Li+ and Na+. Our ICP-MS data demonstrate that there is selectivity between Na+ and Li+ (greater Na+ than Li+ uptake) and, most interestingly, between the Li+ isotopes (greater 6Li+ than 7Li+ uptake) by the inner mitochondrial membrane. In summary, we observed no Li+ isotope differentiation for Ca2+ efflux in mitochondria via NCLX but found a Li+ isotope fractionation during Li+ uptake by mitochondria with NCLX active or blocked. Our results suggest that the transport of Li+ via NCLX is not the main pathway for Li+ isotope fractionation and that this differentiation does not affect Ca2+ efflux in mitochondria. Therefore, explaining the puzzling effects of Li+ isotopes observed in other contexts will require further investigation to identify the molecular targets for Li+ isotope differentiation.
Collapse
Affiliation(s)
- Irina Bukhteeva
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Fasih A. Rahman
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Brian Kendall
- Department of Earth and Environmental Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Robin E. Duncan
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Joe Quadrilatero
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Evgeny V. Pavlov
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| | - Michel J. P. Gingras
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Zoya Leonenko
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
7
|
Wei H, Adelsheim Z, Fischer R, McCarthy MJ. Serum from Myalgic encephalomyelitis/chronic fatigue syndrome patients causes loss of coherence in cellular circadian rhythms. J Neuroimmunol 2023; 381:578142. [PMID: 37393850 PMCID: PMC10527922 DOI: 10.1016/j.jneuroim.2023.578142] [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: 03/01/2023] [Revised: 05/12/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a disabling disorder characterized by disrupted daily patterns of activity, sleep, and physiology. Past studies in ME/CFS patients have examined circadian rhythms, suggested that desynchronization between central and peripheral rhythms may be an important pathological feature, and identified associated changes in post-inflammatory cytokines such as transforming growth factor beta (TGFB). However, no previous studies have examined circadian rhythms in ME/CFS using cellular models or studied the role of cytokines on circadian rhythms. In this study, we used serum samples previously collected from ME/CFS patients (n = 20) selected for the presence of insomnia symptoms and matched controls (n = 20) to determine the effects of serum factors and TGFB on circadian rhythms in NIH3T3 mouse immortalized fibroblasts stably transfected with the Per2-luc bioluminescent circadian reporter. Compared to control serum, ME/CFS serum caused a significant loss of rhythm robustness (decreased goodness of fit) and nominally increased the rate of damping of cellular rhythms. Damping rate was associated with insomnia severity in ME/CFS patients using the Pittsburgh Sleep Quality Index (PSQI). Recombinant TGFB1 peptide applied to cells reduced rhythm amplitude, caused phase delay and decreased robustness of rhythms. However, there was no difference in TGFB1 levels between ME/CFS and control serum indicating the effects of serum on cellular rhythms cannot be explained by levels of this cytokine. Future studies will be required to identify additional serum factors in ME/CFS patients that alter circadian rhythms in cells.
Collapse
Affiliation(s)
- Heather Wei
- VA San Diego Healthcare System, San Diego,CA, USA
| | - Zoe Adelsheim
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Rita Fischer
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Michael J McCarthy
- VA San Diego Healthcare System, San Diego,CA, USA; Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Palm D, Uzoni A, Kronenberg G, Thome J, Faltraco F. Human Derived Dermal Fibroblasts as in Vitro Research Tool to Study Circadian Rhythmicity in Psychiatric Disorders. PHARMACOPSYCHIATRY 2023; 56:87-100. [PMID: 37187177 DOI: 10.1055/a-1147-1552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
A number of psychiatric disorders are defined by persistent or recurrent sleep-wake disturbances alongside disruptions in circadian rhythm and altered clock gene expression. Circadian rhythms are present not only in the hypothalamic suprachiasmatic nucleus but also in peripheral tissues. In this respect, cultures of human derived dermal fibroblasts may serve as a promising new tool to investigate cellular and molecular mechanisms underlying the pathophysiology of mental illness. In this article, we discuss the advantages of fibroblast cultures to study psychiatric disease. More specifically, we provide an update on recent advances in modeling circadian rhythm disorders using human fibroblasts.
Collapse
Affiliation(s)
- Denise Palm
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| | - Adriana Uzoni
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| | - Golo Kronenberg
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| | - Johannes Thome
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| | - Frank Faltraco
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
9
|
Rexrode L, Tennin M, Babu J, Young C, Bollavarapu R, Lawson LA, Valeri J, Pantazopoulos H, Gisabella B. Regulation of dendritic spines in the amygdala following sleep deprivation. FRONTIERS IN SLEEP 2023; 2:1145203. [PMID: 37928499 PMCID: PMC10624159 DOI: 10.3389/frsle.2023.1145203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The amygdala is a hub of emotional circuits involved in the regulation of cognitive and emotional behaviors and its critically involved in emotional reactivity, stress regulation, and fear memory. Growing evidence suggests that the amygdala plays a key role in the consolidation of emotional memories during sleep. Neuroimaging studies demonstrated that the amygdala is selectively and highly activated during rapid eye movement sleep (REM) and sleep deprivation induces emotional instability and dysregulation of the emotional learning process. Regulation of dendritic spines during sleep represents a morphological correlate of memory consolidation. Several studies indicate that dendritic spines are remodeled during sleep, with evidence for broad synaptic downscaling and selective synaptic upscaling in several cortical areas and the hippocampus. Currently, there is a lack of information regarding the regulation of dendritic spines in the amygdala during sleep. In the present work, we investigated the effect of 5 h of sleep deprivation on dendritic spines in the mouse amygdala. Our data demonstrate that sleep deprivation results in differential dendritic spine changes depending on both the amygdala subregions and the morphological subtypes of dendritic spines. We observed decreased density of mushroom spines in the basolateral amygdala of sleep deprived mice, together with increased neck length and decreased surface area and volume. In contrast, we observed greater densities of stubby spines in sleep deprived mice in the central amygdala, indicating that downscaling selectively occurs in this spine type. Greater neck diameters for thin spines in the lateral and basolateral nuclei of sleep deprived mice, and decreases in surface area and volume for mushroom spines in the basolateral amygdala compared to increases in the cental amygdala provide further support for spine type-selective synaptic downscaling in these areas during sleep. Our findings suggest that sleep promotes synaptic upscaling of mushroom spines in the basolateral amygdala, and downscaling of selective spine types in the lateral and central amygdala. In addition, we observed decreased density of phosphorylated cofilin immunoreactive and growth hormone immunoreactive cells in the amygdala of sleep deprived mice, providing further support for upscaling of dendritic spines during sleep. Overall, our findings point to region-and spine type-specific changes in dendritic spines during sleep in the amygdala, which may contribute to consolidation of emotional memories during sleep.
Collapse
Affiliation(s)
- Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Matthew Tennin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jobin Babu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Caleb Young
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lamiorkor Ameley Lawson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, United States
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
10
|
Rohr KE, McCarthy MJ. The impact of lithium on circadian rhythms and implications for bipolar disorder pharmacotherapy. Neurosci Lett 2022; 786:136772. [PMID: 35798199 DOI: 10.1016/j.neulet.2022.136772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/01/2022] [Indexed: 01/21/2023]
Abstract
Bipolar disorder (BD) is characterized by disrupted circadian rhythms affecting sleep, arousal, and mood. Lithium is among the most effective mood stabilizer treatments for BD, and in addition to improving mood symptoms, stabilizes sleep and activity rhythms in treatment responsive patients. Across a variety of experimental models, lithium has effects on circadian rhythms. However, uncertainty exists as to whether these actions directly pertain to lithium's therapeutic effects. Here, we consider evidence from mechanistic studies in animals and cells and clinical trials in BD patients that identify associations between circadian rhythms and the therapeutic effects of lithium. Most evidence indicates that lithium has effects on cellular circadian rhythms and increases morningness behaviors in BD patients, changes that may contribute to the therapeutic effects of lithium. However, much of this evidence is limited by cross-sectional analyses and/or imprecise proxy markers of clinical outcomes and circadian rhythms in BD patients, while mechanistic studies rely on inference from animals or small numbers of patients . Further study may clarify the essential mechanisms underlying lithium responsive BD, better characterize the longitudinal changes in circadian rhythms in BD patients, and inform the development of therapeutic interventions targeting circadian rhythms.
Collapse
Affiliation(s)
- Kayla E Rohr
- Department of Psychiatry and Center For Circadian Biology, University of California San Diego, La Jolla, CA, USA
| | - Michael J McCarthy
- Department of Psychiatry and Center For Circadian Biology, University of California San Diego, La Jolla, CA, USA; Mental Health Service, VA San Diego Healthcare System, La Jolla, CA, USA.
| |
Collapse
|
11
|
McCarthy MJ, Gottlieb JF, Gonzalez R, McClung CA, Alloy LB, Cain S, Dulcis D, Etain B, Frey BN, Garbazza C, Ketchesin KD, Landgraf D, Lee H, Marie‐Claire C, Nusslock R, Porcu A, Porter R, Ritter P, Scott J, Smith D, Swartz HA, Murray G. Neurobiological and behavioral mechanisms of circadian rhythm disruption in bipolar disorder: A critical multi-disciplinary literature review and agenda for future research from the ISBD task force on chronobiology. Bipolar Disord 2022; 24:232-263. [PMID: 34850507 PMCID: PMC9149148 DOI: 10.1111/bdi.13165] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM Symptoms of bipolar disorder (BD) include changes in mood, activity, energy, sleep, and appetite. Since many of these processes are regulated by circadian function, circadian rhythm disturbance has been examined as a biological feature underlying BD. The International Society for Bipolar Disorders Chronobiology Task Force (CTF) was commissioned to review evidence for neurobiological and behavioral mechanisms pertinent to BD. METHOD Drawing upon expertise in animal models, biomarkers, physiology, and behavior, CTF analyzed the relevant cross-disciplinary literature to precisely frame the discussion around circadian rhythm disruption in BD, highlight key findings, and for the first time integrate findings across levels of analysis to develop an internally consistent, coherent theoretical framework. RESULTS Evidence from multiple sources implicates the circadian system in mood regulation, with corresponding associations with BD diagnoses and mood-related traits reported across genetic, cellular, physiological, and behavioral domains. However, circadian disruption does not appear to be specific to BD and is present across a variety of high-risk, prodromal, and syndromic psychiatric disorders. Substantial variability and ambiguity among the definitions, concepts and assumptions underlying the research have limited replication and the emergence of consensus findings. CONCLUSIONS Future research in circadian rhythms and its role in BD is warranted. Well-powered studies that carefully define associations between BD-related and chronobiologically-related constructs, and integrate across levels of analysis will be most illuminating.
Collapse
Affiliation(s)
- Michael J. McCarthy
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
- VA San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - John F. Gottlieb
- Department of PsychiatryFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Robert Gonzalez
- Department of Psychiatry and Behavioral HealthPennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Colleen A. McClung
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lauren B. Alloy
- Department of PsychologyTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sean Cain
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityMelbourneVictoriaAustralia
| | - Davide Dulcis
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | - Bruno Etain
- Université de ParisINSERM UMR‐S 1144ParisFrance
| | - Benicio N. Frey
- Department Psychiatry and Behavioral NeuroscienceMcMaster UniversityHamiltonOntarioCanada
| | - Corrado Garbazza
- Centre for ChronobiologyPsychiatric Hospital of the University of Basel and Transfaculty Research Platform Molecular and Cognitive NeurosciencesUniversity of BaselBaselSwitzerland
| | - Kyle D. Ketchesin
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dominic Landgraf
- Circadian Biology GroupDepartment of Molecular NeurobiologyClinic of Psychiatry and PsychotherapyUniversity HospitalLudwig Maximilian UniversityMunichGermany
| | - Heon‐Jeong Lee
- Department of Psychiatry and Chronobiology InstituteKorea UniversitySeoulSouth Korea
| | | | - Robin Nusslock
- Department of Psychology and Institute for Policy ResearchNorthwestern UniversityChicagoIllinoisUSA
| | - Alessandra Porcu
- UC San Diego Department of Psychiatry & Center for Circadian BiologyLa JollaCaliforniaUSA
| | | | - Philipp Ritter
- Clinic for Psychiatry and PsychotherapyCarl Gustav Carus University Hospital and Technical University of DresdenDresdenGermany
| | - Jan Scott
- Institute of NeuroscienceNewcastle UniversityNewcastleUK
| | - Daniel Smith
- Division of PsychiatryUniversity of EdinburghEdinburghUK
| | - Holly A. Swartz
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Greg Murray
- Centre for Mental HealthSwinburne University of TechnologyMelbourneVictoriaAustralia
| |
Collapse
|
12
|
Abstract
PURPOSE Development of new thymoleptic medications has primarily centered on anticonvulsants and antipsychotic drugs. Based on our studies of intracellular calcium ion signaling in mood disorders, we were interested in the use of novel medications that act on this mechanism of neuronal activation as potential mood stabilizers. METHOD We reviewed the dynamics of the calcium second messenger system and the international body of data demonstrating increased baseline and stimulated intracellular calcium levels in peripheral cells of patients with bipolar mood disorders. We then examined studies of the effect of established mood stabilizers on intracellular calcium ion levels and on mechanisms of mobilization of this second messenger. After summarizing studies of calcium channel blocking agents, whose primary action is to attenuate hyperactive intracellular calcium signaling, we considered clinical experience with this class of medications and the potential for further research. FINDINGS Established mood stabilizers normalize increased intracellular calcium ion levels in bipolar disorder patients. Most case series and controlled studies suggest an antimanic and possibly mood stabilizing effect of the calcium channel blocking medications verapamil and nimodipine, with fewer data on isradipine. A relatively low risk of teratogenicity and lack of cognitive adverse effects or weight gain suggest possible applications in pregnancy and in patients for whom these are considerations. IMPLICATIONS Medications that antagonize hyperactive intracellular signaling warrant more interest than they have received in psychiatry. Further experience will clarify the applications of these medications alone and in combination with more established mood stabilizers.
Collapse
|
13
|
Chronbiologically-based sub-groups in bipolar I disorder: A latent profile analysis. J Affect Disord 2022; 299:691-697. [PMID: 34879259 DOI: 10.1016/j.jad.2021.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Bipolar disorder presents with significant phenotypic heterogeneity. The aim of this study was to investigate whether bipolar disorder, type I (BDI) subjects could be meaningfully classified into homogeneous groups according to activity, sleep, and circadian characteristics using latent profile analysis (LPA). We hypothesized that distinct BDI sub-groups would be identified based primarily on circadian-associated markers. MATERIALS AND METHODS 105 individuals with BDI were included in the study. Seventeen activity, sleep, and circadian characteristics were assessed via actigraphy and clinical assessments. LPA was conducted to stratify our sample into homogenous sub-groups. Differences between groups on demographic, clinical, activity, sleep, and circadian characteristics were explored. RESULTS Two distinct groups were identified, a High Chronobiological Disturbance group (HCD) (56%, N = 59) and a Low Chronobiological Disturbance group (LCD) (41%; N = 46). Circadian variables were the defining characteristics in sub-group determination. Large effect sizes and magnitudes of association were noted in circadian variables between HCD and LCD sub-groups. Several circadian rhythm variables accounted for a large percentage of the variance between HCD and LCD sub-groups. No differences were noted between sub-groups on demographic characteristics and the psychiatric medications currently in use. Mood state did not significantly impact sub-group differences. LIMITATIONS The protocol was cross-sectional in design. Longitudinal studies are required to determine the stability of the identified sub-groups. CONCLUSION LPA was able to identify sub-groups in BDI with circadian variables being the most distinguishing factors in determining sub-group class membership. Future research should explore the role that circadian characteristics can play in defining sub-phenotypes of bipolar disorder.
Collapse
|
14
|
Pisanu C, Congiu D, Severino G, Ardau R, Chillotti C, Del Zompo M, Baune BT, Squassina A. Investigation of genetic loci shared between bipolar disorder and risk-taking propensity: potential implications for pharmacological interventions. Neuropsychopharmacology 2021; 46:1680-1692. [PMID: 34035470 PMCID: PMC8280111 DOI: 10.1038/s41386-021-01045-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 11/09/2022]
Abstract
Patients with bipolar disorder (BD) often show increased risk-taking propensity, which may contribute to poor clinical outcome. While these two phenotypes are genetically correlated, there is scarce knowledge on the shared genetic determinants. Using GWAS datasets on BD (41,917 BD cases and 371,549 controls) and risk-taking (n = 466,571), we dissected shared genetic determinants using conjunctional false discovery rate (conjFDR) and local genetic covariance analysis. We investigated specificity of identified targets using GWAS datasets on schizophrenia (SCZ) and attention-deficit hyperactivity disorder (ADHD). The putative functional role of identified targets was evaluated using different tools and GTEx v. 8. Target druggability was evaluated using DGIdb and enrichment for drug targets with genome for REPositioning drugs (GREP). Among 102 loci shared between BD and risk-taking, 87% showed the same direction of effect. Sixty-two were specifically shared between risk-taking propensity and BD, while the others were also shared between risk-taking propensity and either SCZ or ADHD. By leveraging pleiotropic enrichment, we reported 15 novel and specific loci associated with BD and 22 with risk-taking. Among cross-disorder genes, CACNA1C (a known target of calcium channel blockers) was significantly associated with risk-taking propensity and both BD and SCZ using conjFDR (p = 0.001 for both) as well as local genetic covariance analysis, and predicted to be differentially expressed in the cerebellar hemisphere in an eQTL-informed gene-based analysis (BD, Z = 7.48, p = 3.8E-14; risk-taking: Z = 4.66, p = 1.6E-06). We reported for the first time shared genetic determinants between BD and risk-taking propensity. Further investigation into calcium channel blockers or development of innovative ligands of calcium channels might form the basis for innovative pharmacotherapy in patients with BD with increased risk-taking propensity.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Maria Del Zompo
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
15
|
Xu N, Shinohara K, Saunders KEA, Geddes JR, Cipriani A. Effect of lithium on circadian rhythm in bipolar disorder: A systematic review and meta-analysis. Bipolar Disord 2021; 23:445-453. [PMID: 33650218 DOI: 10.1111/bdi.13070] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/23/2021] [Accepted: 02/21/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Circadian rhythm disruption is commonly reported in patients with bipolar disorder. Lithium has been suggested to have effects on the circadian clock, the biological basis of the circadian rhythm. The objective of the current review was to review systematically the existing studies on the effect of lithium on circadian rhythm in patients with bipolar disorder. METHODS We systematically searched the scientific literature up to September 2020 for experimental or observational studies which measured circadian rhythm in bipolar patients taking lithium (in comparison with placebo or other active treatments) and carried out a meta-analysis. Circadian rest-activity was our primary outcome, but we also collected data about sleep quality and chronotype (Morningness-Eveningness). The protocol was registered in PROSPERO (CRD42018109790). RESULTS Four observational studies (n = 668) and one experimental study (n = 29) were included. Results from the meta-analysis suggest a potential association between lithium and shifts towards morningness (standardized mean difference [SMD]: 0.42, 95% confidence interval [CI]: -0.05 to 0.90). One cohort study with 21 days of follow-up found that patients treated with lithium had significantly larger amplitude (0.68, 0.01 to 1.36) when compared to anticonvulsants. CONCLUSION This review highlights the insufficient evidence to inform us about the effect of lithium on circadian rhythm. However, we found that chronotype can be a potential target for further exploration of biomarkers or biosignatures of lithium treatment in patients with bipolar disorder. Further studies with prospective and longitudinal study design, adopting actigraphy to monitor daily circadian rest-activity changes are needed.
Collapse
Affiliation(s)
- Ni Xu
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Kiyomi Shinohara
- Departmens of Health Promotion and Human Behavior and of Clinical Epidemiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kate E A Saunders
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.,Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.,Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.,Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| |
Collapse
|
16
|
Circadian rhythms in bipolar disorder patient-derived neurons predict lithium response: preliminary studies. Mol Psychiatry 2021; 26:3383-3394. [PMID: 33674753 PMCID: PMC8418615 DOI: 10.1038/s41380-021-01048-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/19/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
Bipolar disorder (BD) is a neuropsychiatric illness defined by recurrent episodes of mania/hypomania, depression and circadian rhythm abnormalities. Lithium is an effective drug for BD, but 30-40% of patients fail to respond adequately to treatment. Previous work has demonstrated that lithium affects the expression of "clock genes" and that lithium responders (Li-R) can be distinguished from non-responders (Li-NR) by differences in circadian rhythms. However, circadian rhythms have not been evaluated in BD patient neurons from Li-R and Li-NR. We used induced pluripotent stem cells (iPSCs) to culture neuronal precursor cells (NPC) and glutamatergic neurons from BD patients characterized for lithium responsiveness and matched controls. We identified strong circadian rhythms in Per2-luc expression in NPCs and neurons from controls and Li-R, but NPC rhythms in Li-R had a shorter circadian period. Li-NR rhythms were low amplitude and profoundly weakened. In NPCs and neurons, expression of PER2 was higher in both BD groups compared to controls. In neurons, PER2 protein levels were higher in BD than controls, especially in Li-NR samples. In single cells, NPC and neuron rhythms in both BD groups were desynchronized compared to controls. Lithium lengthened period in Li-R and control neurons but failed to alter rhythms in Li-NR. In contrast, temperature entrainment increased amplitude across all groups, and partly restored rhythms in Li-NR neurons. We conclude that neuronal circadian rhythm abnormalities are present in BD and most pronounced in Li-NR. Rhythm deficits in BD may be partly reversible through stimulation of entrainment pathways.
Collapse
|
17
|
Gisabella B, Babu J, Valeri J, Rexrode L, Pantazopoulos H. Sleep and Memory Consolidation Dysfunction in Psychiatric Disorders: Evidence for the Involvement of Extracellular Matrix Molecules. Front Neurosci 2021; 15:646678. [PMID: 34054408 PMCID: PMC8160443 DOI: 10.3389/fnins.2021.646678] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances and memory dysfunction are key characteristics across psychiatric disorders. Recent advances have revealed insight into the role of sleep in memory consolidation, pointing to key overlap between memory consolidation processes and structural and molecular abnormalities in psychiatric disorders. Ongoing research regarding the molecular mechanisms involved in memory consolidation has the potential to identify therapeutic targets for memory dysfunction in psychiatric disorders and aging. Recent evidence from our group and others points to extracellular matrix molecules, including chondroitin sulfate proteoglycans and their endogenous proteases, as molecules that may underlie synaptic dysfunction in psychiatric disorders and memory consolidation during sleep. These molecules may provide a therapeutic targets for decreasing strength of reward memories in addiction and traumatic memories in PTSD, as well as restoring deficits in memory consolidation in schizophrenia and aging. We review the evidence for sleep and memory consolidation dysfunction in psychiatric disorders and aging in the context of current evidence pointing to the involvement of extracellular matrix molecules in these processes.
Collapse
Affiliation(s)
| | | | | | | | - Harry Pantazopoulos
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
18
|
Cavieres-Lepe J, Ewer J. Reciprocal Relationship Between Calcium Signaling and Circadian Clocks: Implications for Calcium Homeostasis, Clock Function, and Therapeutics. Front Mol Neurosci 2021; 14:666673. [PMID: 34045944 PMCID: PMC8144308 DOI: 10.3389/fnmol.2021.666673] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/09/2021] [Indexed: 12/03/2022] Open
Abstract
In animals, circadian clocks impose a daily rhythmicity to many behaviors and physiological processes. At the molecular level, circadian rhythms are driven by intracellular transcriptional/translational feedback loops (TTFL). Interestingly, emerging evidence indicates that they can also be modulated by multiple signaling pathways. Among these, Ca2+ signaling plays a key role in regulating the molecular rhythms of clock genes and of the resulting circadian behavior. In addition, the application of in vivo imaging approaches has revealed that Ca2+ is fundamental to the synchronization of the neuronal networks that make up circadian pacemakers. Conversely, the activity of circadian clocks may influence Ca2+ signaling. For instance, several genes that encode Ca2+ channels and Ca2+-binding proteins display a rhythmic expression, and a disruption of this cycling affects circadian function, underscoring their reciprocal relationship. Here, we review recent advances in our understanding of how Ca2+ signaling both modulates and is modulated by circadian clocks, focusing on the regulatory mechanisms described in Drosophila and mice. In particular, we examine findings related to the oscillations in intracellular Ca2+ levels in circadian pacemakers and how they are regulated by canonical clock genes, neuropeptides, and light stimuli. In addition, we discuss how Ca2+ rhythms and their associated signaling pathways modulate clock gene expression at the transcriptional and post-translational levels. We also review evidence based on transcriptomic analyzes that suggests that mammalian Ca2+ channels and transporters (e.g., ryanodine receptor, ip3r, serca, L- and T-type Ca2+ channels) as well as Ca2+-binding proteins (e.g., camk, cask, and calcineurin) show rhythmic expression in the central brain clock and in peripheral tissues such as the heart and skeletal muscles. Finally, we discuss how the discovery that Ca2+ signaling is regulated by the circadian clock could influence the efficacy of pharmacotherapy and the outcomes of clinical interventions.
Collapse
Affiliation(s)
- Javier Cavieres-Lepe
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - John Ewer
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
19
|
Estradiol fluctuations and depressive history as risk factors for menopausal depression. Menopause 2021; 28:234-236. [PMID: 33534431 DOI: 10.1097/gme.0000000000001736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Paul P, Nadella RK, Viswanath B, Reddy YCJ, Jain S, Purushottam M. Does CACNA1C rs1006737 genotype play a role in lithium treatment response in bipolar disorder patients? Asian J Psychiatr 2021; 56:102525. [PMID: 33418281 DOI: 10.1016/j.ajp.2020.102525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Pradip Paul
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Ravi Kumar Nadella
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Biju Viswanath
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Y C Janardhan Reddy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Sanjeev Jain
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Meera Purushottam
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India.
| |
Collapse
|
21
|
Rosenthal SJ, Josephs T, Kovtun O, McCarty R. Seasonal effects on bipolar disorder: A closer look. Neurosci Biobehav Rev 2020; 115:199-219. [DOI: 10.1016/j.neubiorev.2020.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 11/15/2022]
|
22
|
Pisanu C, Congiu D, Manchia M, Caria P, Cocco C, Dettori T, Frau DV, Manca E, Meloni A, Nieddu M, Noli B, Pinna F, Robledo R, Sogos V, Ferri GL, Carpiniello B, Vanni R, Bocchetta A, Severino G, Ardau R, Chillotti C, Zompo MD, Squassina A. Differences in telomere length between patients with bipolar disorder and controls are influenced by lithium treatment. Pharmacogenomics 2020; 21:533-540. [DOI: 10.2217/pgs-2020-0028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: To assess the role of lithium treatment in the relationship between bipolar disorder (BD) and leukocyte telomere length (LTL). Materials & methods: We compared LTL between 131 patients with BD, with or without a history of lithium treatment, and 336 controls. We tested the association between genetically determined LTL and BD in two large genome-wide association datasets. Results: Patients with BD with a history lithium treatment showed longer LTL compared with never-treated patients (p = 0.015), and similar LTL compared with controls. Patients never treated with lithium showed shorter LTL compared with controls (p = 0.029). Mendelian randomization analysis showed no association between BD and genetically determined LTL. Conclusion: Our data support previous findings showing that long-term lithium treatment might protect against telomere shortening.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Donatella Congiu
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Mirko Manchia
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Paola Caria
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Cristina Cocco
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Tinuccia Dettori
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Daniela Virginia Frau
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Elias Manca
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Anna Meloni
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Mariella Nieddu
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Federica Pinna
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Renato Robledo
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Gian Luca Ferri
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Bernardo Carpiniello
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Roberta Vanni
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Alberto Bocchetta
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Giovanni Severino
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Maria Del Zompo
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Alessio Squassina
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| |
Collapse
|
23
|
Abstract
People with bipolar disorder (BD) all too often have suboptimal long-term outcomes with existing treatment options. They experience relapsing episodes of depression and mania and also have interepisodic mood and anxiety symptoms. We need to have a better understanding of the pathophysiology of BD if we are to make progress in improving these outcomes. This chapter will focus on the critical role of mitochondria in human functioning, oxidative stress, and the biological mechanisms of mitochondria in BD. Additionally, this chapter will present the evidence that, at least for some people, BD is a product of mitochondrial dysregulation. We review the modulators of mitochondria, the connection between current BD medication treatments and mitochondria, and additional medications that have theoretical potential to treat BD.
Collapse
|
24
|
Porcu A, Gonzalez R, McCarthy MJ. Pharmacological Manipulation of the Circadian Clock: A Possible Approach to the Management of Bipolar Disorder. CNS Drugs 2019; 33:981-999. [PMID: 31625128 DOI: 10.1007/s40263-019-00673-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bipolar disorder (BD) is a mood disorder with genetic and neurobiological underpinnings, characterized primarily by recurrent episodes of mania and depression, with notable disruptions in rhythmic behaviors such as sleep, energy, appetite and attention. The chronobiological links to BD are further supported by the effectiveness of various treatment modalities such as bright light, circadian phase advance, and mood-stabilizing drugs such as lithium that have effects on the circadian clock. Over the past 30 years, the neurobiology of the circadian clock has been exquisitely described and there now exists a detailed knowledge of key signaling pathways, neurotransmitters and signaling mechanisms that regulate various dimensions of circadian clock function. With this new wealth of information, it is becoming increasingly plausible that new drugs for BD could be made by targeting molecular elements of the circadian clock. However, circadian rhythms are multidimensional and complex, involving unique, time-dependent factors that are not typically considered in drug development. We review the organization of the circadian clock in the central nervous system and briefly summarize data implicating the circadian clock in BD. We then consider some of the unique aspects of the circadian clock as a drug target in BD, discuss key methodological considerations and evaluate some of the candidate clock pathways and systems that could serve as potential targets for novel mood stabilizers. We expect this work will serve as a roadmap to facilitate the development of compounds acting on the circadian clock for the treatment of BD.
Collapse
Affiliation(s)
- Alessandra Porcu
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Robert Gonzalez
- Department of Psychiatry, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, 17033-0850, USA
| | - Michael J McCarthy
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, CA, 92093, USA. .,Psychiatry Service, VA San Diego Healthcare System, 3350 La Jolla Village Dr MC116A, San Diego, CA, 92161, USA.
| |
Collapse
|
25
|
Petersen AS, Barloese MCJ, Snoer A, Soerensen AMS, Jensen RH. Verapamil and Cluster Headache: Still a Mystery. A Narrative Review of Efficacy, Mechanisms and Perspectives. Headache 2019; 59:1198-1211. [DOI: 10.1111/head.13603] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Anja S. Petersen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Mads C. J. Barloese
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging Hvidovre Hospital Hvidovre Denmark
| | - Agneta Snoer
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Anne Mette S. Soerensen
- Department of Clinical Pharmacology Bispebjerg and Frederiksberg Hospital Copenhagen Denmark
| | - Rigmor H. Jensen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| |
Collapse
|
26
|
Prata DP, Costa-Neves B, Cosme G, Vassos E. Unravelling the genetic basis of schizophrenia and bipolar disorder with GWAS: A systematic review. J Psychiatr Res 2019; 114:178-207. [PMID: 31096178 DOI: 10.1016/j.jpsychires.2019.04.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES To systematically review findings of GWAS in schizophrenia (SZ) and in bipolar disorder (BD); and to interpret findings, with a focus on identifying independent replications. METHOD PubMed search, selection and review of all independent GWAS in SZ or BD, published since March 2011, i.e. studies using non-overlapping samples within each article, between articles, and with those of the previous review (Li et al., 2012). RESULTS From the 22 GWAS included in this review, the genetic associations surviving standard GWAS-significance were for genetic markers in the regions of ACSL3/KCNE4, ADCY2, AMBRA1, ANK3, BRP44, DTL, FBLN1, HHAT, INTS7, LOC392301, LOC645434/NMBR, LOC729457, LRRFIP1, LSM1, MDM1, MHC, MIR2113/POU3F2, NDST3, NKAPL, ODZ4, PGBD1, RENBP, TRANK1, TSPAN18, TWIST2, UGT1A1/HJURP, WHSC1L1/FGFR1 and ZKSCAN4. All genes implicated across both reviews are discussed in terms of their function and implication in neuropsychiatry. CONCLUSION Taking all GWAS to date into account, AMBRA1, ANK3, ARNTL, CDH13, EFHD1 (albeit with different alleles), MHC, PLXNA2 and UGT1A1 have been implicated in either disorder in at least two reportedly non-overlapping samples. Additionally, evidence for a SZ/BD common genetic basis is most strongly supported by the implication of ANK3, NDST3, and PLXNA2.
Collapse
Affiliation(s)
- Diana P Prata
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Portugal; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, UK; Instituto Universitário de Lisboa (ISCTE-IUL), Centro de Investigação e Intervenção Social, Lisboa, Portugal.
| | - Bernardo Costa-Neves
- Lisbon Medical School, University of Lisbon, Av. Professor Egas Moniz, 1649-028, Lisbon, Portugal; Centro Hospitalar Psiquiátrico de Lisboa, Av. do Brasil, 53 1749-002, Lisbon, Portugal
| | - Gonçalo Cosme
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Portugal
| | - Evangelos Vassos
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, 16 De Crespigny Park, SE5 8AF, UK
| |
Collapse
|
27
|
Nudell V, Wei H, Nievergelt C, Maihofer AX, Shilling P, Alda M, Berrettini WH, Brennand KJ, Calabrese JR, Coryell WH, Covault JM, Frye MA, Gage F, Gershon E, McInnis MG, Nurnberger JI, Oedegaard KJ, Shekhtman T, Zandi PP, Kelsoe JR, McCarthy MJ. Entrainment of Circadian Rhythms to Temperature Reveals Amplitude Deficits in Fibroblasts from Patients with Bipolar Disorder and Possible Links to Calcium Channels. MOLECULAR NEUROPSYCHIATRY 2019; 5:115-124. [PMID: 31192224 DOI: 10.1159/000497354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/29/2019] [Indexed: 11/19/2022]
Abstract
Bipolar disorder (BD) is characterized by recurrent mood episodes, and circadian rhythm disturbances. Past studies have identified calcium channel genes as risk loci for BD. CACNA1C encodes an L-type calcium channel (LTCC) involved in the entrainment of circadian rhythms to light. Another calcium channel, i.e., the ryanodine receptor (RYR), is involved in -circadian phase delays. It is unknown whether variants in CACNA1C or other calcium channels contribute to the circadian phenotype in BD. We hypothesized that, by using temperature cycles, we could model circadian entrainment in fibroblasts from BD patients and controls to interrogate the circadian functions of LTCCs. Using Per2-luc, a bioluminescent reporter, we verified that cells entrain to temperature rhythms in vitro. Under constant temperature conditions, the LTCC antagonist verapamil shortened the circadian period, and the RYR antagonist dantrolene lengthened the period. However, neither drug affected temperature entrainment. Fibroblasts from BD patients and controls also entrained to temperature. In cells from BD patients, the rhythm amplitude was lower under entrained, but not constant, conditions. Temperature entrainment was otherwise similar between BD and control cells. However, the CACNA1C genotype among BD cells predicted the degree to which cells entrained. We conclude that assessment of rhythms under entrained conditions reveals additional rhythm abnormalities in BD that are not observable under constant temperature conditions.
Collapse
Affiliation(s)
- Victoria Nudell
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Heather Wei
- VA San Diego Healthcare System Psychiatry Service, San Diego, California, USA
| | - Caroline Nievergelt
- VA San Diego Healthcare System Psychiatry Service, San Diego, California, USA.,Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Adam X Maihofer
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Paul Shilling
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Wade H Berrettini
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kristen J Brennand
- Departments of Neuroscience and Psychiatry, Icahn School of Medicine at Mt. Sinai, New York, New York, USA
| | - Joseph R Calabrese
- Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Jonathan M Covault
- Department of Psychiatry, University of Connecticut, Farmington, Connecticut, USA
| | - Mark A Frye
- Department of Psychiatry, Mayo Clinic, Rochester, Minnesota, USA
| | - Fred Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Elliot Gershon
- Department of Psychiatry, University of Chicago, Chicago, Illinois, USA
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - John I Nurnberger
- Department of Psychiatry, Indiana University, Indianapolis, Indiana, USA
| | - Ketil J Oedegaard
- Section for Psychiatry, University of Bergen and NORMENT and KG Jebsen Centre for Neuropsychiatry, Bergen, Norway.,Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Tatyana Shekhtman
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Peter P Zandi
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland, USA
| | - John R Kelsoe
- VA San Diego Healthcare System Psychiatry Service, San Diego, California, USA.,Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| | - Michael J McCarthy
- VA San Diego Healthcare System Psychiatry Service, San Diego, California, USA.,Department of Psychiatry and Center for Circadian Biology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
28
|
McCarthy MJ. Missing a beat: assessment of circadian rhythm abnormalities in bipolar disorder in the genomic era. Psychiatr Genet 2019; 29:29-36. [PMID: 30516584 DOI: 10.1097/ypg.0000000000000215] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Circadian rhythm abnormalities have been recognized as a central feature of bipolar disorder (BD) but a coherent biological explanation for them remains lacking. Using genetic mutation of 'clock genes', robust animal models of mania and depression have been developed that elucidate key aspects of circadian rhythms and the circadian clock-mood connection. However, translation of this knowledge into humans remains incomplete. In recent years, very large genome-wide association studies (GWAS) have been conducted and the genetic underpinnings of BD are beginning to emerge. However, these genetic studies in BD do not match well with the evidence from animal studies that implicate the circadian clock in mood regulation. Even larger GWAS have been conducted for circadian phenotypes including chronotype, rhythm amplitude, sleep duration, and insomnia. These studies have identified a diverse set of associated genes, including a minority with previously well-characterized functions in the circadian clock. Taken together, the data from recent GWAS of BD and circadian phenotypes indicate that the genetic organization of the circadian clock, both in health and in BD is complex. The findings from GWAS elucidate potentially novel circadian mechanism that may be partly distinct from those identified in animal models. Pleiotropy, epistasis and nongenetic factors may play important roles in regulating circadian rhythms, some of which may underlie circadian rhythm disturbances in BD.
Collapse
Affiliation(s)
- Michael J McCarthy
- Department of Psychiatry, Center for Circadian Biology, VA San Diego Healthcare System, University of California San Diego, San Diego, California, USA
| |
Collapse
|
29
|
SNAP-25 in Major Psychiatric Disorders: A Review. Neuroscience 2019; 420:79-85. [PMID: 30790667 DOI: 10.1016/j.neuroscience.2019.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 01/10/2019] [Accepted: 02/07/2019] [Indexed: 02/07/2023]
Abstract
Synaptosomal Associated Protein-25 kilodaltons (SNAP-25) is an integral member of the SNARE complex. This complex is essential for calcium-triggered synaptic vesicular fusion and release of neurotransmitters into the synaptic cleft. In addition to neurotransmission, SNAP-25 is associated with insulin release, the regulation of intracellular calcium, and neuroplasticity. Because of SNAP-25's varied and crucial biological roles, the consequences of changes in this protein can be seen in both the central nervous system and the periphery. In this review, we will look at the published literature from human genetic, postmortem, and animal studies involving SNAP-25. The accumulated data indicate that SNAP-25 may be linked with some symptoms associated with a variety of psychiatric disorders. These disorders include bipolar disorder, schizophrenia, major depressive disorder, attention deficit hyperactivity disorder, autism, alcohol use disorder, and dementia. There are also data suggesting SNAP-25 may be involved with non-psychiatric seizures and metabolic disorders. We believe investigation of SNAP-25 is important for understanding both normal behavior and some aspects of the pathophysiology of behavior seen with psychiatric disorders. The wealth of information from both animal and human studies on SNAP-25 offers an excellent opportunity to use a bi-directional research approach. Hypotheses generated from genetically manipulated mice can be directly tested in human postmortem tissue, and, conversely, human genetic and postmortem findings can improve and validate animal models for psychiatric disorders.
Collapse
|
30
|
McCarthy MJ, Wei H, Nievergelt CM, Stautland A, Maihofer AX, Welsh DK, Shilling P, Alda M, Alliey-Rodriguez N, Anand A, Andreasson OA, Balaraman Y, Berrettini WH, Bertram H, Brennand KJ, Calabrese JR, Calkin CV, Claasen A, Conroy C, Coryell WH, Craig DW, D’Arcangelo N, Demodena A, Djurovic S, Feeder S, Fisher C, Frazier N, Frye MA, Gage FH, Gao K, Garnham J, Gershon ES, Glazer K, Goes F, Goto T, Harrington G, Jakobsen P, Kamali M, Karberg E, Kelly M, Leckband SG, Lohoff F, McInnis MG, Mondimore F, Morken G, Nurnberger JI, Obral S, Oedegaard KJ, Ortiz A, Ritchey M, Ryan K, Schinagle M, Schoeyen H, Schwebel C, Shaw M, Shekhtman T, Slaney C, Stapp E, Szelinger S, Tarwater B, Zandi PP, Kelsoe JR. Chronotype and cellular circadian rhythms predict the clinical response to lithium maintenance treatment in patients with bipolar disorder. Neuropsychopharmacology 2019; 44:620-628. [PMID: 30487653 PMCID: PMC6333516 DOI: 10.1038/s41386-018-0273-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a serious mood disorder associated with circadian rhythm abnormalities. Risk for BD is genetically encoded and overlaps with systems that maintain circadian rhythms. Lithium is an effective mood stabilizer treatment for BD, but only a minority of patients fully respond to monotherapy. Presently, we hypothesized that lithium-responsive BD patients (Li-R) would show characteristic differences in chronotype and cellular circadian rhythms compared to lithium non-responders (Li-NR). Selecting patients from a prospective, multi-center, clinical trial of lithium monotherapy, we examined morning vs. evening preference (chronotype) as a dimension of circadian rhythm function in 193 Li-R and Li-NR BD patients. From a subset of 59 patient donors, we measured circadian rhythms in skin fibroblasts longitudinally over 5 days using a bioluminescent reporter (Per2-luc). We then estimated circadian rhythm parameters (amplitude, period, phase) and the pharmacological effects of lithium on rhythms in cells from Li-R and Li-NR donors. Compared to Li-NRs, Li-Rs showed a difference in chronotype, with higher levels of morningness. Evening chronotype was associated with increased mood symptoms at baseline, including depression, mania, and insomnia. Cells from Li-Rs were more likely to exhibit a short circadian period, a linear relationship between period and phase, and period shortening effects of lithium. Common genetic variation in the IP3 signaling pathway may account for some of the individual differences in the effects of lithium on cellular rhythms. We conclude that circadian rhythms may influence response to lithium in maintenance treatment of BD.
Collapse
Affiliation(s)
- Michael J. McCarthy
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA ,0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Heather Wei
- 0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Caroline M. Nievergelt
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA
| | - Andrea Stautland
- 0000 0004 1936 7443grid.7914.bDepartment of Clinical Medicine, Section for Psychiatry, University of Bergen, Bergen, Norway
| | - Adam X. Maihofer
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA
| | - David K. Welsh
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA ,0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Paul Shilling
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA
| | - Martin Alda
- 0000 0004 1936 8200grid.55602.34Department of Psychiatry, Dalhousie University, Halifax, Canada
| | - Ney Alliey-Rodriguez
- 0000 0004 1936 7822grid.170205.1Department of Psychiatry, University of Chicago, Chicago, USA
| | - Amit Anand
- 0000 0001 0790 959Xgrid.411377.7Department of Psychiatry, Indiana University, Bloomington, USA
| | - Ole A. Andreasson
- 0000 0004 1936 8921grid.5510.1Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
| | - Yokesh Balaraman
- 0000 0001 0790 959Xgrid.411377.7Department of Psychiatry, Indiana University, Bloomington, USA
| | - Wade H. Berrettini
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Holli Bertram
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Kristen J. Brennand
- 0000 0001 0670 2351grid.59734.3cDepartments of Neuroscience and Psychiatry, Icahn School of Medicine at Mt Sinai, New York, USA
| | - Joseph R. Calabrese
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Cynthia V. Calkin
- 0000 0004 1936 8200grid.55602.34Department of Psychiatry, Dalhousie University, Halifax, Canada
| | - Ana Claasen
- 0000 0004 0507 3225grid.250942.8Neurogenomics Division, Translational Genomics Research Institute, Phoenix, USA
| | - Clara Conroy
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - William H. Coryell
- 0000 0004 1936 8294grid.214572.7Department of Psychiatry, University of Iowa, Iowa City, USA
| | - David W. Craig
- 0000 0004 0507 3225grid.250942.8Neurogenomics Division, Translational Genomics Research Institute, Phoenix, USA
| | - Nicole D’Arcangelo
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Anna Demodena
- 0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Srdjan Djurovic
- 0000 0004 1936 8921grid.5510.1Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
| | - Scott Feeder
- 0000 0004 0459 167Xgrid.66875.3aDepartment of Psychiatry, The Mayo Clinic, Rochester, USA
| | - Carrie Fisher
- 0000 0001 0790 959Xgrid.411377.7Department of Psychiatry, Indiana University, Bloomington, USA
| | - Nicole Frazier
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Mark A. Frye
- 0000 0004 0459 167Xgrid.66875.3aDepartment of Psychiatry, The Mayo Clinic, Rochester, USA
| | - Fred H. Gage
- 0000 0001 0662 7144grid.250671.7Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, USA
| | - Keming Gao
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Julie Garnham
- 0000 0004 1936 8200grid.55602.34Department of Psychiatry, Dalhousie University, Halifax, Canada
| | - Elliot S. Gershon
- 0000 0004 1936 7822grid.170205.1Department of Psychiatry, University of Chicago, Chicago, USA
| | - Kara Glazer
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - Fernando Goes
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - Toyomi Goto
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Gloria Harrington
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Petter Jakobsen
- 0000 0000 9753 1393grid.412008.fNorment and KG Jebsen Centre for Neuropsychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Masoud Kamali
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Elizabeth Karberg
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Marisa Kelly
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Susan G. Leckband
- 0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Falk Lohoff
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Melvin G. McInnis
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Francis Mondimore
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - Gunnar Morken
- 0000 0001 1516 2393grid.5947.fDivision of Psychiatry, St. Olav University Hospital of Trondheim and Department of Mental Health Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - John I. Nurnberger
- 0000 0001 0790 959Xgrid.411377.7Department of Psychiatry, Indiana University, Bloomington, USA
| | - Sarah Obral
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Ketil J. Oedegaard
- 0000 0004 1936 7443grid.7914.bDepartment of Clinical Medicine, Section for Psychiatry, University of Bergen, Bergen, Norway ,0000 0000 9753 1393grid.412008.fNorment and KG Jebsen Centre for Neuropsychiatry, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Abigail Ortiz
- 0000 0001 2182 2255grid.28046.38Department of Psychiatry, University of Ottawa, Ottawa, ON Canada
| | - Megan Ritchey
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - Kelly Ryan
- 0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Martha Schinagle
- 0000 0001 2164 3847grid.67105.35Department of Psychiatry, Case Western Reserve University, Cleveland, OH USA
| | - Helle Schoeyen
- 0000 0004 1936 7443grid.7914.bDepartment of Clinical Medicine, Section for Psychiatry, University of Bergen, Bergen, Norway
| | - Candice Schwebel
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Martha Shaw
- 0000 0004 1936 7822grid.170205.1Department of Psychiatry, University of Chicago, Chicago, USA
| | - Tatyana Shekhtman
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA ,0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| | - Claire Slaney
- 0000 0004 1936 8200grid.55602.34Department of Psychiatry, Dalhousie University, Halifax, Canada
| | - Emma Stapp
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - Szabolcs Szelinger
- 0000 0004 0507 3225grid.250942.8Neurogenomics Division, Translational Genomics Research Institute, Phoenix, USA
| | - Bruce Tarwater
- 0000 0004 0507 3225grid.250942.8Neurogenomics Division, Translational Genomics Research Institute, Phoenix, USA
| | - Peter P. Zandi
- 0000 0001 2171 9311grid.21107.35Department of Psychiatry, Johns Hopkins University, Baltimore, USA
| | - John R. Kelsoe
- 0000 0001 2107 4242grid.266100.3Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, 3350 La Jolla Village Dr. MC 116A, San Diego, CA 92161 USA ,0000 0004 0419 2708grid.410371.0Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161 USA
| |
Collapse
|
31
|
Abstract
A large series of different ion channels have been identified and investigated as potential targets for new medicines for the treatment of a variety of human diseases, including pain. Among these channels, the voltage gated calcium channels (VGCC) are inhibited by drugs for the treatment of migraine, neuropathic pain or intractable pain. Transient receptor potential (TRP) channels are emerging as important pain transducers as they sense low pH media or oxidative stress and other mediators and are abundantly found at sites of inflammation or tissue injury. Low pH may also activate acid sensing ion channels (ASIC) and mechanical forces stimulate the PIEZO channels. While potent agonists of TRP channels due to their desensitizing action on pain transmission are used as topical applications, the potential of TRP antagonists as pain therapeutics remains an exciting field of investigation. The study of ASIC or PIEZO channels in pain signaling is in an early stage, whereas antagonism of the purinergic P2X3 channels has been reported to provide beneficial effects in chronic intractable cough. The present chapter covers these intriguing channels in great detail, highlighting their diverse mechanisms and broad potential for therapeutic utility.
Collapse
Affiliation(s)
- Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| |
Collapse
|
32
|
Dubovsky SL. Applications of calcium channel blockers in psychiatry: pharmacokinetic and pharmacodynamic aspects of treatment of bipolar disorder. Expert Opin Drug Metab Toxicol 2018; 15:35-47. [PMID: 30558453 DOI: 10.1080/17425255.2019.1558206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Calcium channel blockers (CCBs) comprise a heterogeneous group of medications that reduce calcium influx and attenuate cellular hyperactivity. Evidence of hyperactive intracellular calcium ion signaling in multiple peripheral cells of patients with bipolar disorder, calcium antagonist actions of established mood stabilizers, and a relative dearth of treatments have prompted research into potential uses of CCBs for this common and disabling condition. Areas covered: This review provides a comprehensive overview of intracellular calcium signaling in bipolar disorder, structure and function of calcium channels, pharmacology of CCBs, evidence of efficacy of CCBs in bipolar disorder, clinical applications, and directions for future research. Expert opinion: Despite mixed evidence of efficacy, CCBs are a promising novel approach to a demonstrated cellular abnormality in both poles of bipolar disorder. Potential advantages include low potential for sedation and weight gain, and possible usefulness for pregnant and neurologically impaired patients. Further research should focus on markers of a preferential response, studies in specific bipolar subtypes, development of CCBs acting preferentially in the central nervous system and on calcium channels that are primarily involved in neuronal signaling and plasticity.
Collapse
Affiliation(s)
- Steven L Dubovsky
- a Department of Psychiatry , State University of New York at Buffalo , Buffalo , NY , USA.,b Departments of Psychiatry and Medicine , University of Colorado , Denver , CO , USA
| |
Collapse
|
33
|
Kelly RM, Healy U, Sreenan S, McDermott JH, Coogan AN. Clocks in the clinic: circadian rhythms in health and disease. Postgrad Med J 2018; 94:653-658. [PMID: 30523071 DOI: 10.1136/postgradmedj-2018-135719] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/30/2018] [Accepted: 11/17/2018] [Indexed: 11/03/2022]
Abstract
Circadian rhythms are endogenously generated recurring patterns of around 24 hours with well-established roles in physiology and behaviour. These circadian clocks are important in both the aetiology and treatment of various psychiatric and metabolic diseases. To maintain physiological homeostasis and optimal functioning, living life synchronised to these clocks is desirable; modern society, however, promotes a '24/7' lifestyle where activity often occurs during the body's 'biological night', resulting in mistimed sleep and circadian misalignment. This circadian desynchrony can increase the risk of disease and can also influence treatment response. Clinicians should be aware of the influence that circadian desynchrony can have on health and disease, in order to potentially develop new therapeutic strategies and to incorporate chronotherapeutics into current treatment strategies to enhance their utility.
Collapse
Affiliation(s)
- Rachael M Kelly
- Department of Psychology, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Ultan Healy
- Academic Department of Endocrinology, Royal College of Surgeons in Ireland, Connolly Hospital Blanchardstown, Dublin, Ireland.,3U Diabetes Consortium, Dublin, Ireland
| | - Seamus Sreenan
- Academic Department of Endocrinology, Royal College of Surgeons in Ireland, Connolly Hospital Blanchardstown, Dublin, Ireland.,3U Diabetes Consortium, Dublin, Ireland
| | - John H McDermott
- Academic Department of Endocrinology, Royal College of Surgeons in Ireland, Connolly Hospital Blanchardstown, Dublin, Ireland.,3U Diabetes Consortium, Dublin, Ireland
| | - Andrew N Coogan
- Department of Psychology, Maynooth University, National University of Ireland, Maynooth, Ireland
| |
Collapse
|
34
|
Chua CEL, Tang BL. miR-34a in Neurophysiology and Neuropathology. J Mol Neurosci 2018; 67:235-246. [DOI: 10.1007/s12031-018-1231-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022]
|
35
|
Wei H, Landgraf D, Wang G, McCarthy MJ. Inositol polyphosphates contribute to cellular circadian rhythms: Implications for understanding lithium's molecular mechanism. Cell Signal 2018; 44:82-91. [PMID: 29331582 DOI: 10.1016/j.cellsig.2018.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/08/2017] [Accepted: 01/07/2018] [Indexed: 12/21/2022]
Abstract
Most living organisms maintain cell autonomous circadian clocks that synchronize critical biological functions with daily environmental cycles. In mammals, the circadian clock is regulated by inputs from signaling pathways including glycogen synthase kinase 3 (GSK3). The drug lithium has actions on GSK3, and also on inositol metabolism. While it is suspected that lithium's inhibition of GSK3 causes rhythm changes, it is not known if inositol polyphosphates can also affect the circadian clock. We examined whether the signaling molecule inositol hexaphosphate (IP6) has effects on circadian rhythms. Using a bioluminescent reporter (Per2::luc) to measure circadian rhythms, we determined that IP6 increased rhythm amplitude and shortened period in NIH3T3 cells. The IP6 effect on amplitude was attenuated by selective siRNA knockdown of GSK3B and pharmacological blockade of AKT kinase. However, unlike lithium, IP6 did not induce serine-9 phosphorylation of GSK3B. The synthesis of IP6 involves the enzymes inositol polyphosphate multikinase (IPMK) and inositol pentakisphosphate 2-kinase (IPPK). Knockdown of Ippk had effects opposite to those of IP6, decreasing rhythm amplitude and lengthening period. Ipmk knockdown had few effects on rhythm alone, but attenuated the effects of lithium on rhythms. However, lithium did not change the intracellular content of IP6 in NIH3T3 cells or neurons. Pharmacological inhibition of the IP6 kinases (IP6K) increased rhythm amplitude and shortened period, suggesting secondary effects of inositol pyrophosphates may underlie the period shortening effect, but not the amplitude increasing effect of IP6. Overall, we conclude that inositol phosphates, in particular IP6 have effects on circadian rhythms. Manipulations affecting IP6 and related inositol phosphates may offer a novel means through which circadian rhythms can be regulated.
Collapse
Affiliation(s)
- Heather Wei
- Research and Psychiatry Service, Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA 92161, United States; Department of Psychiatry and Center for Circadian Biology, University of California San Diego, 9500 Gilman Dr La Jolla, CA 92093, United States
| | - Dominic Landgraf
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, 9500 Gilman Dr La Jolla, CA 92093, United States
| | - George Wang
- Department of Psychiatry and Center for Circadian Biology, University of California San Diego, 9500 Gilman Dr La Jolla, CA 92093, United States
| | - Michael J McCarthy
- Research and Psychiatry Service, Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA 92161, United States; Department of Psychiatry and Center for Circadian Biology, University of California San Diego, 9500 Gilman Dr La Jolla, CA 92093, United States.
| |
Collapse
|
36
|
Circadian Rhythm Disturbances in Mood Disorders: Insights into the Role of the Suprachiasmatic Nucleus. Neural Plast 2017; 2017:1504507. [PMID: 29230328 PMCID: PMC5694588 DOI: 10.1155/2017/1504507] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/05/2017] [Accepted: 10/03/2017] [Indexed: 12/28/2022] Open
Abstract
Circadian rhythm disturbances are a common symptom among individuals with mood disorders. The suprachiasmatic nucleus (SCN), in the ventral part of the anterior hypothalamus, orchestrates physiological and behavioral circadian rhythms. The SCN consists of self-sustaining oscillators and receives photic and nonphotic cues, which entrain the SCN to the external environment. In turn, through synaptic and hormonal mechanisms, the SCN can drive and synchronize circadian rhythms in extra-SCN brain regions and peripheral tissues. Thus, genetic or environmental perturbations of SCN rhythms could disrupt brain regions more closely related to mood regulation and cause mood disturbances. Here, we review clinical and preclinical studies that provide evidence both for and against a causal role for the SCN in mood disorders.
Collapse
|
37
|
Melo MC, Abreu RL, Linhares Neto VB, de Bruin PF, de Bruin VM. Chronotype and circadian rhythm in bipolar disorder: A systematic review. Sleep Med Rev 2017; 34:46-58. [DOI: 10.1016/j.smrv.2016.06.007] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 12/01/2022]
|
38
|
Harrison PJ, Cipriani A, Harmer CJ, Nobre AC, Saunders K, Goodwin GM, Geddes JR. Innovative approaches to bipolar disorder and its treatment. Ann N Y Acad Sci 2017; 1366:76-89. [PMID: 27111134 PMCID: PMC4850752 DOI: 10.1111/nyas.13048] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 12/29/2022]
Abstract
All psychiatric disorders have suffered from a dearth of truly novel pharmacological interventions. In bipolar disorder, lithium remains a mainstay of treatment, six decades since its effects were serendipitously discovered. The lack of progress reflects several factors, including ignorance of the disorder's pathophysiology and the complexities of the clinical phenotype. After reviewing the current status, we discuss some ways forward. First, we highlight the need for a richer characterization of the clinical profile, facilitated by novel devices and new forms of data capture and analysis; such data are already promoting a reevaluation of the phenotype, with an emphasis on mood instability rather than on discrete clinical episodes. Second, experimental medicine can provide early indications of target engagement and therapeutic response, reducing the time, cost, and risk involved in evaluating potential mood stabilizers. Third, genomic data can inform target identification and validation, such as the increasing evidence for involvement of calcium channel genes in bipolar disorder. Finally, new methods and models relevant to bipolar disorder, including stem cells and genetically modified mice, are being used to study key pathways and drug effects. A combination of these approaches has real potential to break the impasse and deliver genuinely new treatments.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Catherine J Harmer
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Anna C Nobre
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom.,Oxford Centre for Human Brain Activity, Warneford Hospital, Oxford, United Kingdom
| | - Kate Saunders
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Guy M Goodwin
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
39
|
Cronin P, McCarthy MJ, Lim ASP, Salmon DP, Galasko D, Masliah E, De Jager PL, Bennett DA, Desplats P. Circadian alterations during early stages of Alzheimer's disease are associated with aberrant cycles of DNA methylation in BMAL1. Alzheimers Dement 2017; 13:689-700. [PMID: 27883893 PMCID: PMC5785929 DOI: 10.1016/j.jalz.2016.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/16/2016] [Accepted: 10/10/2016] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Circadian alterations are prevalent in Alzheimer's disease (AD) and may contribute to cognitive impairment, behavioral symptoms, and neurodegeneration. Epigenetic mechanisms regulate the circadian clock, and changes in DNA methylation have been reported in AD brains, but the pathways that mediate circadian deregulation in AD are incompletely understood. We hypothesized that aberrant DNA methylation may affect circadian rhythms in AD. METHODS We investigated DNA methylation, transcription, and expression of BMAL1, a positive regulator of the circadian clock, in cultured fibroblasts and brain samples from two independent cohorts of aging and AD. RESULTS DNA methylation modulated rhythmic expression of clock genes in cultured fibroblasts. Moreover, rhythmic methylation of BMAL1 was altered in AD brains and fibroblasts and correlated with transcription cycles. DISCUSSION Our results indicate that cycles of DNA methylation contribute to the regulation of BMAL1 rhythms in the brain. Hence, aberrant epigenetic patterns may be linked to circadian alterations in AD.
Collapse
Affiliation(s)
- Peter Cronin
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Michael J McCarthy
- Psychiatry Service, Veterans Affairs San Diego Health Care System, San Diego, CA, USA; Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Andrew S P Lim
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - David P Salmon
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Philip L De Jager
- Departments of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Paula Desplats
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
40
|
Higgins GA, Allyn-Feuer A, Georgoff P, Nikolian V, Alam HB, Athey BD. Mining the topography and dynamics of the 4D Nucleome to identify novel CNS drug pathways. Methods 2017; 123:102-118. [PMID: 28385536 DOI: 10.1016/j.ymeth.2017.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The pharmacoepigenome can be defined as the active, noncoding province of the genome including canonical spatial and temporal regulatory mechanisms of gene regulation that respond to xenobiotic stimuli. Many psychotropic drugs that have been in clinical use for decades have ill-defined mechanisms of action that are beginning to be resolved as we understand the transcriptional hierarchy and dynamics of the nucleus. In this review, we describe spatial, temporal and biomechanical mechanisms mediated by psychotropic medications. Focus is placed on a bioinformatics pipeline that can be used both for detection of pharmacoepigenomic variants that discretize drug response and adverse events to improve pharmacogenomic testing, and for the discovery of novel CNS therapeutics. This approach integrates the functional topology and dynamics of the transcriptional hierarchy of the pharmacoepigenome, gene variant-driven identification of pharmacogenomic regulatory domains, and mesoscale mapping for the discovery of novel CNS pharmacodynamic pathways in human brain. Examples of the application of this pipeline are provided, including the discovery of valproic acid (VPA) mediated transcriptional reprogramming of neuronal cell fate following injury, and mapping of a CNS pathway glutamatergic pathway for the mood stabilizer lithium. These examples in regulatory pharmacoepigenomics illustrate how ongoing research using the 4D nucleome provides a foundation to further insight into previously unrecognized psychotropic drug pharmacodynamic pathways in the human CNS.
Collapse
Affiliation(s)
- Gerald A Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Patrick Georgoff
- Department of Surgery, University of Michigan Medical School, USA
| | - Vahagn Nikolian
- Department of Surgery, University of Michigan Medical School, USA
| | - Hasan B Alam
- Department of Surgery, University of Michigan Medical School, USA
| | - Brian D Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA; Michigan Institute for Data Science (MIDAS), USA.
| |
Collapse
|
41
|
Amare AT, Schubert KO, Klingler-Hoffmann M, Cohen-Woods S, Baune BT. The genetic overlap between mood disorders and cardiometabolic diseases: a systematic review of genome wide and candidate gene studies. Transl Psychiatry 2017; 7:e1007. [PMID: 28117839 PMCID: PMC5545727 DOI: 10.1038/tp.2016.261] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/21/2016] [Accepted: 10/31/2016] [Indexed: 12/11/2022] Open
Abstract
Meta-analyses of genome-wide association studies (meta-GWASs) and candidate gene studies have identified genetic variants associated with cardiovascular diseases, metabolic diseases and mood disorders. Although previous efforts were successful for individual disease conditions (single disease), limited information exists on shared genetic risk between these disorders. This article presents a detailed review and analysis of cardiometabolic diseases risk (CMD-R) genes that are also associated with mood disorders. First, we reviewed meta-GWASs published until January 2016, for the diseases 'type 2 diabetes, coronary artery disease, hypertension' and/or for the risk factors 'blood pressure, obesity, plasma lipid levels, insulin and glucose related traits'. We then searched the literature for published associations of these CMD-R genes with mood disorders. We considered studies that reported a significant association of at least one of the CMD-R genes and 'depression' or 'depressive disorder' or 'depressive symptoms' or 'bipolar disorder' or 'lithium treatment response in bipolar disorder', or 'serotonin reuptake inhibitors treatment response in major depression'. Our review revealed 24 potential pleiotropic genes that are likely to be shared between mood disorders and CMD-Rs. These genes include MTHFR, CACNA1D, CACNB2, GNAS, ADRB1, NCAN, REST, FTO, POMC, BDNF, CREB, ITIH4, LEP, GSK3B, SLC18A1, TLR4, PPP1R1B, APOE, CRY2, HTR1A, ADRA2A, TCF7L2, MTNR1B and IGF1. A pathway analysis of these genes revealed significant pathways: corticotrophin-releasing hormone signaling, AMPK signaling, cAMP-mediated or G-protein coupled receptor signaling, axonal guidance signaling, serotonin or dopamine receptors signaling, dopamine-DARPP32 feedback in cAMP signaling, circadian rhythm signaling and leptin signaling. Our review provides insights into the shared biological mechanisms of mood disorders and cardiometabolic diseases.
Collapse
Affiliation(s)
- A T Amare
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - K O Schubert
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Northern Adelaide Local Health Network, Mental Health Services, Adelaide, SA, Australia
| | - M Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - S Cohen-Woods
- School of Psychology, Faculty of Social and Behavioural Sciences, Flinders University, Adelaide, SA, Australia
| | - B T Baune
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Discipline of Psychiatry, School of Medicine, The University of Adelaide, North Terrace, Adelaide, SA 5005, Australia. E-mail:
| |
Collapse
|
42
|
Malhi GS, Outhred T. Therapeutic Mechanisms of Lithium in Bipolar Disorder: Recent Advances and Current Understanding. CNS Drugs 2016; 30:931-49. [PMID: 27638546 DOI: 10.1007/s40263-016-0380-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lithium is the most effective and well established treatment for bipolar disorder, and it has a broad array of effects within cellular pathways. However, the specific processes through which therapeutic effects occur and are maintained in bipolar disorder remain unclear. This paper provides a timely update to an authoritative review of pertinent findings that was published in CNS Drugs in 2013. A literature search was conducted using the Scopus database, and was limited by year (from 2012). There has been a resurgence of interest in lithium therapy mechanisms, perhaps driven by technical advancements in recent years that permit the examination of cellular mechanisms underpinning the effects of lithium-along with the reuptake of lithium in clinical practice. Recent research has further cemented glycogen synthase kinase 3β (GSK3β) inhibition as a key mechanism, and the inter-associations between GSK3β-mediated neuroprotective, anti-oxidative and neurotransmission mechanisms have been further elucidated. In addition to highly illustrative cellular research, studies examining higher-order biological systems, such as circadian rhythms, as well as employing innovative animal and human models, have increased our understanding of how lithium-induced changes at the cellular level possibly translate to changes at behavioural and clinical levels. Neural circuitry research is yet to identify clear mechanisms of change in bipolar disorder in response to treatment with lithium, but important structural findings have demonstrated links to the modulation of cellular mechanisms, and peripheral marker and pharmacogenetic studies are showing promising findings that will likely inform the exploration for predictors of lithium treatment response. With a deeper understanding of lithium's therapeutic mechanisms-from the cellular to clinical levels of investigation-comes the opportunity to develop predictive models of lithium treatment response and identify novel drug targets, and recent findings have provided important leads towards these goals.
Collapse
Affiliation(s)
- Gin S Malhi
- Academic Department of Psychiatry, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia. .,Sydney Medical School Northern, The University of Sydney, Sydney, NSW, 2006, Australia. .,CADE Clinic Level 3, Main Hospital Building, Royal North Shore Hospital, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia.
| | - Tim Outhred
- Academic Department of Psychiatry, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia.,Sydney Medical School Northern, The University of Sydney, Sydney, NSW, 2006, Australia.,CADE Clinic Level 3, Main Hospital Building, Royal North Shore Hospital, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
| |
Collapse
|
43
|
Cipriani A, Saunders K, Attenburrow MJ, Stefaniak J, Panchal P, Stockton S, Lane TA, Tunbridge EM, Geddes JR, Harrison PJ. A systematic review of calcium channel antagonists in bipolar disorder and some considerations for their future development. Mol Psychiatry 2016; 21:1324-32. [PMID: 27240535 PMCID: PMC5030455 DOI: 10.1038/mp.2016.86] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/01/2016] [Accepted: 04/15/2016] [Indexed: 12/17/2022]
Abstract
l-type calcium channel (LTCC) antagonists have been used in bipolar disorder for over 30 years, without becoming an established therapeutic approach. Interest in this class of drugs has been rekindled by the discovery that LTCC genes are part of the genetic aetiology of bipolar disorder and related phenotypes. We have therefore conducted a systematic review of LTCC antagonists in the treatment and prophylaxis of bipolar disorder. We identified 23 eligible studies, with six randomised, double-blind, controlled clinical trials, all of which investigated verapamil in acute mania, and finding no evidence that it is effective. Data for other LTCC antagonists (diltiazem, nimodipine, nifedipine, methyoxyverapamil and isradipine) and for other phases of the illness are limited to observational studies, and therefore no robust conclusions can be drawn. Given the increasingly strong evidence for calcium signalling dysfunction in bipolar disorder, the therapeutic candidacy of this class of drugs has become stronger, and hence we also discuss issues relevant to their future development and evaluation. In particular, we consider how genetic, molecular and pharmacological data can be used to improve the selectivity, efficacy and tolerability of LTCC antagonists. We suggest that a renewed focus on LTCCs as targets, and the development of 'brain-selective' LTCC ligands, could be one fruitful approach to innovative pharmacotherapy for bipolar disorder and related phenotypes.
Collapse
Affiliation(s)
- A Cipriani
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - K Saunders
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - M-J Attenburrow
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - J Stefaniak
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - P Panchal
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - S Stockton
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - T A Lane
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - E M Tunbridge
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - J R Geddes
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - P J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| |
Collapse
|
44
|
Safari R, Salimi R, Tunca Z, Ozerdem A, Ceylan D, Sakizli M. Mutation/SNP analysis in EF-hand calcium binding domain of mitochondrial Ca2+ uptake 1 gene in bipolar disorder patients. J Integr Neurosci 2016; 15:163-73. [DOI: 10.1142/s0219635216500096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
45
|
Moreira J, Geoffroy PA. Lithium and bipolar disorder: Impacts from molecular to behavioural circadian rhythms. Chronobiol Int 2016; 33:351-73. [DOI: 10.3109/07420528.2016.1151026] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
Kálmán S, Garbett KA, Janka Z, Mirnics K. Human dermal fibroblasts in psychiatry research. Neuroscience 2016; 320:105-21. [PMID: 26855193 DOI: 10.1016/j.neuroscience.2016.01.067] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
In order to decipher the disease etiology, progression and treatment of multifactorial human brain diseases we utilize a host of different experimental models. Recently, patient-derived human dermal fibroblast (HDF) cultures have re-emerged as promising in vitro functional system for examining various cellular, molecular, metabolic and (patho)physiological states and traits of psychiatric disorders. HDF studies serve as a powerful complement to postmortem and animal studies, and often appear to be informative about the altered homeostasis in neural tissue. Studies of HDFs from patients with schizophrenia (SZ), depression, bipolar disorder (BD), autism, attention deficit and hyperactivity disorder and other psychiatric disorders have significantly advanced our understanding of these devastating diseases. These reports unequivocally prove that signal transduction, redox homeostasis, circadian rhythms and gene*environment (G*E) interactions are all amenable for assessment by the HDF model. Furthermore, the reported findings suggest that this underutilized patient biomaterial, combined with modern molecular biology techniques, may have both diagnostic and prognostic value, including prediction of response to therapeutic agents.
Collapse
Affiliation(s)
- S Kálmán
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary.
| | - K A Garbett
- Department of Psychiatry, Vanderbilt University, 8128 MRB III, 465 21st Avenue, Nashville, TN 37232, USA.
| | - Z Janka
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary.
| | - K Mirnics
- Department of Psychiatry, University of Szeged, 57 Kálvária Sgt, Szeged 6725, Hungary; Department of Psychiatry, Vanderbilt University, 8128 MRB III, 465 21st Avenue, Nashville, TN 37232, USA.
| |
Collapse
|