1
|
Berenjian A, Bakhtiarizadeh MR, Mohammadi-Sangcheshmeh A, Sharifi SD. A nutrigenomics approach to study the effects of ω-3 fatty acids in laying hens under physiological stress. Front Physiol 2023; 14:1198247. [PMID: 37560158 PMCID: PMC10407228 DOI: 10.3389/fphys.2023.1198247] [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: 04/05/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Supplement of ω-3 fatty acids can decrease the harmful effects of stress. However, the potential molecular mechanisms that are modulated by dietary ω-3 fatty acids in laying hens under stress remain unknown. Hence, RNA-sequencing (RNA-Seq) technology was used to gain new insights into different gene expression profiles and potential pathways involved in response to stress in the liver of 35-week-old Lohmann LSL-Lite laying hens supplemented with ω-3. Three groups including control (non-stress), stress, and stress_ω-3 fatty acids (three layers per each group) were applied. A total of 1,321 genes were detected as differentially expressed genes of which 701, 1,049, and 86 DEGs belonged to stress vs. control, stress_ω-3 vs. control, and stress vs. stress_ω-3 pairwise comparisons, respectively. Gene ontology and KEGG pathway analysis indicated that the DEGs were enriched in particular regulation of steroid and cholesterol biosynthetic process, fatty acid degradation, AMPK signaling pathway, fatty acid biosynthesis, and immune response. Our data represented a promising approach regarding the importance of ω-3 as anxiolytic and anti-stress. In this context, UNC13B and ADRA1B genes were downregulated in the stress_ω-3 group compared to the stress group, which are associated with decreased activity of glutamatergic stimulatory neurons and probably play important role in facilitating the response to stress. This study extends the current understanding of the liver transcriptome response to physiological stress, and provides new insights into the molecular responses to stress in laying hens fed a diet supplemented with ω-3 fatty acids.
Collapse
Affiliation(s)
| | | | | | - Seyed Davood Sharifi
- Department of Animal and Poultry Science, Faculty of Agricultural Technology, College of Agriculture and Natural Resources, University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Carvajal F, Sánchez-Gil A, Cardona D, Rincón-Cervera MA, Lerma-Cabrera JM. The Effect of Very-Long-Chain n-3 Polyunsaturated Fatty Acids in the Central Nervous System and Their Potential Benefits for Treating Alcohol Use Disorder: Reviewing Pre-Clinical and Clinical Data. Nutrients 2023; 15:2993. [PMID: 37447319 DOI: 10.3390/nu15132993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Alcohol use poses a significant global health concern, leading to serious physical and socioeconomic issues worldwide. The current treatment options for problematic alcohol consumption are limited, leading to the exploration of alternative approaches, such as nutraceuticals. One promising target is very-long-chain n-3 polyunsaturated fatty acids (VLC n-3 PUFAs). This review aims to compile the most relevant pre-clinical and clinical evidence on the effect of VLC n-3 PUFAs on alcohol use disorders and related outcomes. The findings suggest that VLC n-3 PUFAs may alleviate the physiological changes induced by alcohol consumption, including neuroinflammation and neurotransmitter dysregulation. Additionally, they can reduce withdrawal symptoms, improve mood, and reduce stress level, all of which are closely associated with problematic alcohol consumption. However, more research is required to fully understand the precise mechanisms by which VLC n-3 PUFAs exert their function. Furthermore, PUFAs should not be considered a standalone solution, but as a complement to other therapeutic approaches. Although preliminary evidence supports the potential therapeutic effect of VLC n-3 PUFAs on problematic alcohol consumption, additional research is needed to validate these findings and determine the optimal use of PUFAs as part of a comprehensive approach to the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Francisca Carvajal
- Department of Psychology, University of Almeria, 04120 Almeria, Spain
- Health Research Center, University of Almeria, 04120 Almeria, Spain
| | - Ainhoa Sánchez-Gil
- Department of Psychology, University of Almeria, 04120 Almeria, Spain
- Health Research Center, University of Almeria, 04120 Almeria, Spain
| | - Diana Cardona
- Health Research Center, University of Almeria, 04120 Almeria, Spain
- Department of Nursing, Physiotherapy and Medicine, University of Almeria, 04120 Almeria, Spain
| | - Miguel Angel Rincón-Cervera
- Food Technology Division, ceiA3, CIAMBITAL, University of Almeria, 04120 Almeria, Spain
- Institute of Nutrition and Food Technology, University of Chile, Santiago 830490, Chile
| | - Jose Manuel Lerma-Cabrera
- Department of Psychology, University of Almeria, 04120 Almeria, Spain
- Health Research Center, University of Almeria, 04120 Almeria, Spain
| |
Collapse
|
3
|
Bhagar R, Le-Niculescu H, Roseberry K, Kosary K, Daly C, Ballew A, Yard M, Sandusky GE, Niculescu AB. Temporal effects on death by suicide: empirical evidence and possible molecular correlates. DISCOVER MENTAL HEALTH 2023; 3:10. [PMID: 37861857 PMCID: PMC10501025 DOI: 10.1007/s44192-023-00035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/01/2023] [Indexed: 10/21/2023]
Abstract
Popular culture and medical lore have long postulated a connection between full moon and exacerbations of psychiatric disorders. We wanted to empirically analyze the hypothesis that suicides are increased during the period around full moons. We analyzed pre-COVID suicides from the Marion County Coroner's Office (n = 776), and show that deaths by suicide are significantly increased during the week of the full moon (p = 0.037), with older individuals (age ≥ 55) showing a stronger effect (p = 0.019). We also examined in our dataset which hour of the day (3-4 pm, p = 0.035), and which month of the year (September, p = 0.09) show the most deaths by suicide. We had blood samples on a subset of the subjects (n = 45), which enabled us to look at possible molecular mechanisms. We tested a list of top blood biomarkers for suicidality (n = 154) from previous studies of ours 7, to assess which of them are predictive. The biomarkers for suicidality that are predictive of death by suicide during full moon, peak hour of day, and peak month of year, respectively, compared to outside of those periods, appear to be enriched in circadian clock genes. For full moon it is AHCYL2, ACSM3, AK2, and RBM3. For peak hour it is GSK3B, AK2, and PRKCB. For peak month it is TBL1XR1 and PRKCI. Half of these genes are modulated in expression by lithium and by valproate in opposite direction to suicidality, and all of them are modulated by depression and alcohol in the same direction as suicidality. These data suggest that there are temporal effects on suicidality, possibly mediated by biological clocks, pointing to changes in ambient light (timing and intensity) as a therapeutically addressable target to decrease suicidality, that can be coupled with psychiatric pharmacological and addiction treatment preventive interventions.
Collapse
Affiliation(s)
- R Bhagar
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA
| | - K Roseberry
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA
| | - K Kosary
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA
| | - C Daly
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA
| | - A Ballew
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - M Yard
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - G E Sandusky
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 W. 15thStreet, Indianapolis, IN, 46202, USA.
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indianapolis VA Medical Center, Indianapolis, USA.
| |
Collapse
|
4
|
Zhang S, Lu Y, Shi W, Ren Y, Xiao K, Chen W, Li L, Zhao J. SIRT1/FOXO1 Axis-Mediated Hippocampal Angiogenesis is Involved in the Antidepressant Effect of Chaihu Shugan San. Drug Des Devel Ther 2022; 16:2783-2801. [PMID: 36039087 PMCID: PMC9419814 DOI: 10.2147/dddt.s370825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
Affiliation(s)
- Shan Zhang
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Yujia Lu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Wei Shi
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Yi Ren
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Kaihui Xiao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Li Li
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
| | - Jingjie Zhao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
- Department of Integrated Traditional and Western Medicine, Capital Medical University, Beijing, 100050, People’s Republic of China
- Correspondence: Jingjie Zhao, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Beijing, 100050, People’s Republic of China, Tel/Fax +86 10-63139096, Email
| |
Collapse
|
5
|
Stoychev K, Dilkov D, Naghavi E, Kamburova Z. Genetic Basis of Dual Diagnosis: A Review of Genome-Wide Association Studies (GWAS) Focusing on Patients with Mood or Anxiety Disorders and Co-Occurring Alcohol-Use Disorders. Diagnostics (Basel) 2021; 11:1055. [PMID: 34201295 PMCID: PMC8228390 DOI: 10.3390/diagnostics11061055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Comorbidity between Alcohol Use Disorders (AUD), mood, and anxiety disorders represents a significant health burden, yet its neurobiological underpinnings are elusive. The current paper reviews all genome-wide association studies conducted in the past ten years, sampling patients with AUD and co-occurring mood or anxiety disorder(s). (2) Methods: In keeping with PRISMA guidelines, we searched EMBASE, Medline/PUBMED, and PsycINFO databases (January 2010 to December 2020), including references of enrolled studies. Study selection was based on predefined criteria and data underwent a multistep revision process. (3) Results: 15 studies were included. Some of them explored dual diagnoses phenotypes directly while others employed correlational analysis based on polygenic risk score approach. Their results support the significant overlap of genetic factors involved in AUDs and mood and anxiety disorders. Comorbidity risk seems to be conveyed by genes engaged in neuronal development, connectivity, and signaling although the precise neuronal pathways and mechanisms remain unclear. (4) Conclusion: given that genes associated with complex traits including comorbid clinical presentations are of small effect, and individually responsible for a very low proportion of the total variance, larger samples consisting of multiple refined comorbid combinations and confirmed by re-sequencing approaches will be necessary to disentangle the genetic architecture of dual diagnosis.
Collapse
Affiliation(s)
- Kaloyan Stoychev
- Department of Psychiatry, Medical University Pleven, 5800 Pleven, Bulgaria
| | - Dancho Dilkov
- Department of Psychiatry, Military Medical Academy Sofia, 1606 Sofia, Bulgaria;
| | | | - Zornitsa Kamburova
- Department of Medical Genetics, Medical University Pleven, 5800 Pleven, Bulgaria;
| |
Collapse
|
6
|
Jašarević E, Hecht PM, Fritsche KL, Geary DC, Rivera RM, Beversdorf DQ. Maternal DHA supplementation influences sex-specific disruption of placental gene expression following early prenatal stress. Biol Sex Differ 2021; 12:10. [PMID: 33422127 PMCID: PMC7797134 DOI: 10.1186/s13293-020-00356-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/26/2020] [Indexed: 01/06/2023] Open
Abstract
Early life adversity is widely recognized as a key risk factor for early developmental perturbations and contributes to the presentation of neuropsychiatric disorders in adulthood. Neurodevelopmental disorders exhibit a strong sex bias in susceptibility, presentation, onset, and severity, although the underlying mechanisms conferring vulnerability are not well understood. Environmental perturbations during pregnancy, such as malnutrition or stress, have been associated with sex-specific reprogramming that contribute to increased disease risk in adulthood, whereby stress and nutritional insufficiency may be additive and further exacerbate poor offspring outcomes. To determine whether maternal supplementation of docosahexanoic acid (DHA) exerts an effect on offspring outcome following exposure to early prenatal stress (EPS), dams were fed a purified 10:1 omega-6/omega-3 diet supplemented with either 1.0% preformed DHA/kg feed weight (DHA-enriched) or no additional DHA (denoted as the control diet, CTL). Dams were administered chronic variable stress during the first week of pregnancy (embryonic day, E0.5–7.5), and developmental milestones were assessed at E 12.5. Exposure to early prenatal stress (EPS) decreased placenta and embryo weight in males, but not females, exposed to the CTL diet. DHA enrichment reversed the sex-specific decrease in placenta and embryo weight following EPS. Early prenatal exposure upregulated expression of genes associated with oxygen and nutrient transport, including hypoxia inducible factor 3α (HIF3α), peroxisome proliferator-activated receptor alpha (PPARα), and insulin-like growth binding factor 1 (IGFBP1), in the placenta of CTL diet males exposed to EPS. DHA enrichment in EPS-exposed animals abrogated the male-specific upregulation of PPARα, HIF3α, and IGFBP1. Taken together, these studies suggest that maternal dietary DHA enrichment may buffer against maternal stress programming of sex-specific outcomes during early development.
Collapse
Affiliation(s)
- Eldin Jašarević
- Interdisciplinary Neuroscience Program, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA.,Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA
| | - Patrick M Hecht
- Interdisciplinary Neuroscience Program, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA.,Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA
| | - David C Geary
- Interdisciplinary Neuroscience Program, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA.,Department of Psychological Sciences, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA
| | - Rocío M Rivera
- Division of Animal Sciences, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA
| | - David Q Beversdorf
- Interdisciplinary Neuroscience Program, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA. .,Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA. .,Department of Psychological Sciences, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA. .,Department of Radiology, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA. .,Department of Neurology, University of Missouri, One Hospital Drive, DC069.10, Columbia, MO, 65211, USA.
| |
Collapse
|
7
|
Zhang Z, Chen G. A logical relationship for schizophrenia, bipolar, and major depressive disorder. Part 1: Evidence from chromosome 1 high density association screen. J Comp Neurol 2020; 528:2620-2635. [PMID: 32266715 DOI: 10.1002/cne.24921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
Familial clustering of schizophrenia (SCZ), bipolar disorder (BPD), and major depressive disorder (MDD) was investigated systematically (Aukes et al., Genetics in Medicine, 2012, 14, 338-341) and any two or even three of these disorders could coexist in some families. Furthermore, evidence from symptomatology and psychopharmacology also imply the existence of intrinsic connections between these three major psychiatric disorders. A total of 71,445 SNPs on chromosome 1 were genotyped on 119 SCZ, 253 BPD (type-I), 177 MDD cases and 1000 controls and further validated in 986 SCZ patients in the population of Shandong province of China. Outstanding psychosis genes are systematically revealed( ATP1A4, ELTD1, FAM5C, HHAT, KIF26B, LMX1A, NEGR1, NFIA, NR5A2, NTNG1, PAPPA2, PDE4B, PEX14, RYR2, SYT6, TGFBR3, TTLL7, and USH2A). Unexpectedly, flanking genes for up to 97.09% of the associated SNPs were also replicated in an enlarged cohort of 986 SCZ patients. From the perspective of etiological rather than clinical psychiatry, bipolar, and major depressive disorder could be subtypes of schizophrenia. Meanwhile, the varied clinical feature and prognosis might be the result of interaction of genetics and epigenetics, for example, irreversible or reversible shut down, and over or insufficient expression of certain genes, which may gives other aspects of these severe mental disorders.
Collapse
Affiliation(s)
- Zhihua Zhang
- Shandong Mental Health Center, Jinan, Shandong, China
| | - Gang Chen
- Department of Medical Genetics, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
8
|
Le-Niculescu H, Roseberry K, Levey DF, Rogers J, Kosary K, Prabha S, Jones T, Judd S, McCormick MA, Wessel AR, Williams A, Phalen PL, Mamdani F, Sequeira A, Kurian SM, Niculescu AB. Towards precision medicine for stress disorders: diagnostic biomarkers and targeted drugs. Mol Psychiatry 2020; 25:918-938. [PMID: 30862937 PMCID: PMC7192849 DOI: 10.1038/s41380-019-0370-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 01/09/2023]
Abstract
The biological fingerprint of environmental adversity may be key to understanding health and disease, as it encompasses the damage induced as well as the compensatory reactions of the organism. Metabolic and hormonal changes may be an informative but incomplete window into the underlying biology. We endeavored to identify objective blood gene expression biomarkers for psychological stress, a subjective sensation with biological roots. To quantify the stress perception at a particular moment in time, we used a simple visual analog scale for life stress in psychiatric patients, a high-risk group. Then, using a stepwise discovery, prioritization, validation, and testing in independent cohort design, we were successful in identifying gene expression biomarkers that were predictive of high-stress states and of future psychiatric hospitalizations related to stress, more so when personalized by gender and diagnosis. One of the top biomarkers that survived discovery, prioritization, validation, and testing was FKBP5, a well-known gene involved in stress response, which serves as a de facto reassuring positive control. We also compared our biomarker findings with telomere length (TL), another well-established biological marker of psychological stress and show that newly identified predictive biomarkers such as NUB1, APOL3, MAD1L1, or NKTR are comparable or better state or trait predictors of stress than TL or FKBP5. Over half of the top predictive biomarkers for stress also had prior evidence of involvement in suicide, and the majority of them had evidence in other psychiatric disorders, providing a molecular underpinning for the effects of stress in those disorders. Some of the biomarkers are targets of existing drugs, of potential utility in patient stratification, and pharmacogenomics approaches. Based on our studies and analyses, the biomarkers with the best overall convergent functional evidence (CFE) for involvement in stress were FKBP5, DDX6, B2M, LAIR1, RTN4, and NUB1. Moreover, the biomarker gene expression signatures yielded leads for possible new drug candidates and natural compounds upon bioinformatics drug repurposing analyses, such as calcium folinate and betulin. Our work may lead to improved diagnosis and treatment for stress disorders such as PTSD, that result in decreased quality of life and adverse outcomes, including addictions, violence, and suicide.
Collapse
Affiliation(s)
- H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Roseberry
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Rogers
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Kosary
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Prabha
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T Jones
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - S Judd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M A McCormick
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A R Wessel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Williams
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - P L Phalen
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - F Mamdani
- Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA, USA
| | - A Sequeira
- Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA, USA
| | - S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indianapolis VA Medical Center, Indianapolis, IN, USA.
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Galduróz JCF, Bezerra AG, Pires GN, Pauluci R, Noto AR. OMEGA-3 Interventions in Alcohol Dependence and Related Outcomes: A Systematic Review and Propositions. Curr Neuropharmacol 2020; 18:456-462. [PMID: 31989899 PMCID: PMC7457439 DOI: 10.2174/1570159x18666200128120729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/29/2019] [Accepted: 01/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pharmacological treatment for alcohol dependence has only three approved drugs: disulfiram, naltrexone and acamprosate. The effects of these drugs are, however, limited, presenting several side effects and a modestly higher efficacy compared to placebo. The administration of omega-3 might bring new perspectives to relapse prevention. METHODS This systematic review aimed to analyze the available literature, compiling the studies that used omega-3 to prevent relapse in alcohol dependents. RESULTS The databases used were PubMed and Web of Science. We identified 2,231 studies and only five articles addressed the administration of omega-3 and alcoholism. Preclinical studies evaluating the effects of PUFAs related to chronic alcohol administration showed improvements in behavioral, cellular and molecular levels. The clinical trial yielded inconclusive results. CONCLUSION Despite the reduced number of studies, omega-3 interventions seem to be promising for controlling issues related to alcohol dependence.
Collapse
Affiliation(s)
- José Carlos F. Galduróz
- Address correspondence to this author at the Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil;, Tel: +55 11 2149-0155; E-mail:
| | | | | | | | | |
Collapse
|
10
|
Veldic M, Millischer V, Port JD, Ho AMC, Jia YF, Geske JR, Biernacka JM, Backlund L, McElroy SL, Bond DJ, Villaescusa JC, Skime M, Choi DS, Lavebratt C, Schalling M, Frye MA. Genetic variant in SLC1A2 is associated with elevated anterior cingulate cortex glutamate and lifetime history of rapid cycling. Transl Psychiatry 2019; 9:149. [PMID: 31123248 PMCID: PMC6533282 DOI: 10.1038/s41398-019-0483-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/07/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
Glutamatergic dysregulation is implicated in the neurobiology of mood disorders. This study investigated the relationship between the anterior cingulate cortex (AC) glutamate, as measured by proton magnetic resonance spectroscopy (1H-MRS), and single-nucleotide polymorphisms (SNPs) from four genes (GLUL, SLC1A3, SLC1A2, and SLC1A7) that regulate the extracellular glutamate in 26 depressed patients with major depressive disorder (MDD; n = 15) and bipolar disorder (BD; n = 11). Two SNPs (rs3812778 and rs3829280), in perfect linkage disequilibrium, in the 3' untranslated region of the EAAT2 gene SLC1A2, were associated with AC glutamate, with minor allele carriers having significantly higher glutamate levels (p < 0.001) in comparison with common allele homozygotes. In silico analysis revealed an association of minor allele carriers of rs3812778/rs382920 with an upregulation of the astrocytic marker CD44 localized downstream of SLC1A2 on chromosome 11. Finally, we tested the disease relevance of these SNPs in a large group of depressed patients [MDD (n = 458); BD (n = 1473)] and found that minor allele carriers had a significantly higher risk for rapid cycling (p = 0.006). Further work is encouraged to delineate the functional impact of excitatory amino acid transporter genetic variation on CD44 associated physiology and glutamatergic neurotransmission, specifically glutamate-glutamine cycling, and its contribution to subphenotypes of mood disorders.
Collapse
Affiliation(s)
- Marin Veldic
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA.
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - John D Port
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Ada Man-Choi Ho
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Yun-Fang Jia
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jennifer R Geske
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Joanna M Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Lena Backlund
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Susan L McElroy
- Lindner Center of Hope, University of Cincinnati, Cincinnati, OH, USA
| | - David J Bond
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - J Carlos Villaescusa
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
11
|
Towards precision medicine for pain: diagnostic biomarkers and repurposed drugs. Mol Psychiatry 2019; 24:501-522. [PMID: 30755720 PMCID: PMC6477790 DOI: 10.1038/s41380-018-0345-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/30/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
We endeavored to identify objective blood biomarkers for pain, a subjective sensation with a biological basis, using a stepwise discovery, prioritization, validation, and testing in independent cohorts design. We studied psychiatric patients, a high risk group for co-morbid pain disorders and increased perception of pain. For discovery, we used a powerful within-subject longitudinal design. We were successful in identifying blood gene expression biomarkers that were predictive of pain state, and of future emergency department (ED) visits for pain, more so when personalized by gender and diagnosis. MFAP3, which had no prior evidence in the literature for involvement in pain, had the most robust empirical evidence from our discovery and validation steps, and was a strong predictor for pain in the independent cohorts, particularly in females and males with PTSD. Other biomarkers with best overall convergent functional evidence for involvement in pain were GNG7, CNTN1, LY9, CCDC144B, and GBP1. Some of the individual biomarkers identified are targets of existing drugs. Moreover, the biomarker gene expression signatures were used for bioinformatic drug repurposing analyses, yielding leads for possible new drug candidates such as SC-560 (an NSAID), and amoxapine (an antidepressant), as well as natural compounds such as pyridoxine (vitamin B6), cyanocobalamin (vitamin B12), and apigenin (a plant flavonoid). Our work may help mitigate the diagnostic and treatment dilemmas that have contributed to the current opioid epidemic.
Collapse
|
12
|
Wolstenholme JT, Bowers MS, Pais AB, Pais AC, Poland RS, Poklis JL, Davies AG, Bettinger JC. Dietary Omega-3 Fatty Acids Differentially Impact Acute Ethanol-Responsive Behaviors and Ethanol Consumption in DBA/2J Versus C57BL/6J Mice. Alcohol Clin Exp Res 2018; 42:1476-1485. [PMID: 29786878 DOI: 10.1111/acer.13780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/15/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Complex interactions between environmental and genetic factors influence the risk of developing alcohol use disorder (AUD) in humans. To date, studies of the impact of environment on AUD risk have primarily focused on psychological characteristics or on the effects of developmental exposure to ethanol (EtOH). We recently observed that modifying levels of the long-chain ω-3 (LC ω-3) fatty acid, eicosapentaenoic acid (EPA), alters acute physiological responses to EtOH in Caenorhabditis elegans. Because mammals derive ω-3 fatty acids from their diet, here we asked if manipulating dietary levels of LC ω-3 fatty acids can affect EtOH-responsive behaviors in mice. METHODS We used 2 well-characterized inbred mouse strains, C57BL/6J (B6) and DBA/2J (D2), which differ in their responses to EtOH. Age-matched young adult male mice were maintained on isocaloric diets that differed only by being enriched or depleted in LC ω-3 fatty acids. Animals were subsequently tested for acute EtOH sensitivity (locomotor activation and sedation), voluntary consumption, and metabolism. Fat deposition was also determined. RESULTS We found that dietary levels of LC ω-3s altered EtOH sensitivity and consumption in a genotype-specific manner. Both B6 and D2 animals fed high LC ω-3 diets demonstrated lower EtOH-induced locomotor stimulation than those fed low LC ω-3 diets. EtOH sedation and EtOH metabolism were greater in D2, but not B6 mice on the high LC ω-3 diet. Conversely, LC ω-3 dietary manipulation altered EtOH consumption in B6, but not in D2 mice. B6 mice on a high LC ω-3 diet consumed more EtOH in a 2-bottle choice intermittent access model than B6 mice on a low LC ω-3 diet. CONCLUSIONS Because EtOH sensitivity is predictive of risk of developing AUD in humans, our data indicate that dietary LC ω-3 levels should be evaluated for their impact on AUD risk in humans. Further, these studies indicate that genetic background can interact with fatty acids in the diet to significantly alter EtOH-responsive behaviors.
Collapse
Affiliation(s)
- Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia.,VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| | - M Scott Bowers
- Department of Biomedical Engineering , Faulk Center for Molecular Therapeutics, Northwestern University, Chicago, Illinois
| | - Alexander B Pais
- VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| | - A Christian Pais
- VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan S Poland
- VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| | - Justin L Poklis
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia
| | - Andrew G Davies
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia.,VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| | - Jill C Bettinger
- Department of Pharmacology and Toxicology , Virginia Commonwealth University, Richmond, Virginia.,VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
13
|
Teng S, Thomson PA, McCarthy S, Kramer M, Muller S, Lihm J, Morris S, Soares DC, Hennah W, Harris S, Camargo LM, Malkov V, McIntosh AM, Millar JK, Blackwood DH, Evans KL, Deary IJ, Porteous DJ, McCombie WR. Rare disruptive variants in the DISC1 Interactome and Regulome: association with cognitive ability and schizophrenia. Mol Psychiatry 2018; 23:1270-1277. [PMID: 28630456 PMCID: PMC5984079 DOI: 10.1038/mp.2017.115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
Schizophrenia (SCZ), bipolar disorder (BD) and recurrent major depressive disorder (rMDD) are common psychiatric illnesses. All have been associated with lower cognitive ability, and show evidence of genetic overlap and substantial evidence of pleiotropy with cognitive function and neuroticism. Disrupted in schizophrenia 1 (DISC1) protein directly interacts with a large set of proteins (DISC1 Interactome) that are involved in brain development and signaling. Modulation of DISC1 expression alters the expression of a circumscribed set of genes (DISC1 Regulome) that are also implicated in brain biology and disorder. Here we report targeted sequencing of 59 DISC1 Interactome genes and 154 Regulome genes in 654 psychiatric patients and 889 cognitively-phenotyped control subjects, on whom we previously reported evidence for trait association from complete sequencing of the DISC1 locus. Burden analyses of rare and singleton variants predicted to be damaging were performed for psychiatric disorders, cognitive variables and personality traits. The DISC1 Interactome and Regulome showed differential association across the phenotypes tested. After family-wise error correction across all traits (FWERacross), an increased burden of singleton disruptive variants in the Regulome was associated with SCZ (FWERacross P=0.0339). The burden of singleton disruptive variants in the DISC1 Interactome was associated with low cognitive ability at age 11 (FWERacross P=0.0043). These results identify altered regulation of schizophrenia candidate genes by DISC1 and its core Interactome as an alternate pathway for schizophrenia risk, consistent with the emerging effects of rare copy number variants associated with intellectual disability.
Collapse
Affiliation(s)
- S Teng
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Biology, Howard University, Washington DC, USA
| | - P A Thomson
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - S McCarthy
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - M Kramer
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - S Muller
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - J Lihm
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - S Morris
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - D C Soares
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - W Hennah
- Institute for Molecular Medicine, Finland FIMM, University of Helsinki, Helsinki, Finland
| | - S Harris
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - L M Camargo
- UCB New Medicines, One Broadway, Cambridge, MA, USA
| | - V Malkov
- Genetics and Pharmacogenomics, MRL, Merck & Co, Boston, MA, USA
| | - A M McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - J K Millar
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - D H Blackwood
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - K L Evans
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - I J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - D J Porteous
- Centre for Genomic and Experimental Medicine, MRC/University of Edinburgh Institute of Genetics & Molecular Medicine, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - W R McCombie
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
14
|
Xiao HW, Ge C, Feng GX, Li Y, Luo D, Dong JL, Li H, Wang H, Cui M, Fan SJ. Gut microbiota modulates alcohol withdrawal-induced anxiety in mice. Toxicol Lett 2018; 287:23-30. [PMID: 29391279 DOI: 10.1016/j.toxlet.2018.01.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
Excessive alcohol consumption remains a major public health problem that affects millions of people worldwide. Accumulative experimental evidence has suggested an important involvement of gut microbiota in the modulation of host's immunological and neurological functions. However, it is previously unknown whether enteric microbiota is implicated in the formation of alcohol withdrawal-induced anxiety. Using a murine model of chronic alcoholism and withdrawal, we examined the impact of alcohol consumption on the possible alterations of gut microbiota as well as alcohol withdrawal-induced anxiety and behavior changes. The 16S rRNA sequencing revealed that alcohol consumption did not alter the abundance of bacteria, but markedly changed the composition of gut microbiota. Moreover, the transplantation of enteric microbes from alcohol-fed mice to normal healthy controls remarkably shaped the composition of gut bacteria, and elicited behavioral signs of alcohol withdrawal-induced anxiety. Using quantitative real-time polymerase chain reaction, we further confirmed that the expression of genes implicated in alcohol addiction, BDNF, CRHR1 and OPRM1, was also altered by transplantation of gut microbes from alcohol-exposed donors. Collectively, our findings suggested a possibility that the alterations of gut microbiota composition might contribute to the development of alcohol withdrawal-induced anxiety, and reveal potentially new etiologies for treating alcohol addiction.
Collapse
Affiliation(s)
- Hui-Wen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Chang Ge
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Guo-Xing Feng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Dan Luo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Jia-Li Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China
| | - Haichao Wang
- Department of Emergency Medicine, North Shore University Hospital, Laboratory of Emergency Medicine, the Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China.
| | - Sai-Jun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin 300192, China.
| |
Collapse
|
15
|
Niculescu AB, Le-Niculescu H, Levey DF, Phalen PL, Dainton HL, Roseberry K, Niculescu EM, Niezer JO, Williams A, Graham DL, Jones TJ, Venugopal V, Ballew A, Yard M, Gelbart T, Kurian SM, Shekhar A, Schork NJ, Sandusky GE, Salomon DR. Precision medicine for suicidality: from universality to subtypes and personalization. Mol Psychiatry 2017; 22:1250-1273. [PMID: 28809398 PMCID: PMC5582166 DOI: 10.1038/mp.2017.128] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 01/15/2023]
Abstract
Suicide remains a clear, present and increasing public health problem, despite being a potentially preventable tragedy. Its incidence is particularly high in people with overt or un(der)diagnosed psychiatric disorders. Objective and precise identification of individuals at risk, ways of monitoring response to treatments and novel preventive therapeutics need to be discovered, employed and widely deployed. We sought to investigate whether blood gene expression biomarkers for suicide (that is, a 'liquid biopsy' approach) can be identified that are more universal in nature, working across psychiatric diagnoses and genders, using larger cohorts than in previous studies. Such markers may reflect and/or be a proxy for the core biology of suicide. We were successful in this endeavor, using a comprehensive stepwise approach, leading to a wealth of findings. Steps 1, 2 and 3 were discovery, prioritization and validation for tracking suicidality, resulting in a Top Dozen list of candidate biomarkers comprising the top biomarkers from each step, as well as a larger list of 148 candidate biomarkers that survived Bonferroni correction in the validation step. Step 4 was testing the Top Dozen list and Bonferroni biomarker list for predictive ability for suicidal ideation (SI) and for future hospitalizations for suicidality in independent cohorts, leading to the identification of completely novel predictive biomarkers (such as CLN5 and AK2), as well as reinforcement of ours and others previous findings in the field (such as SLC4A4 and SKA2). Additionally, we examined whether subtypes of suicidality can be identified based on mental state at the time of high SI and identified four potential subtypes: high anxiety, low mood, combined and non-affective (psychotic). Such subtypes may delineate groups of individuals that are more homogenous in terms of suicidality biology and behavior. We also studied a more personalized approach, by psychiatric diagnosis and gender, with a focus on bipolar males, the highest risk group. Such a personalized approach may be more sensitive to gender differences and to the impact of psychiatric co-morbidities and medications. We compared testing the universal biomarkers in everybody versus testing by subtypes versus personalized by gender and diagnosis, and show that the subtype and personalized approaches permit enhanced precision of predictions for different universal biomarkers. In particular, LHFP appears to be a strong predictor for suicidality in males with depression. We also directly examined whether biomarkers discovered using male bipolars only are better predictors in a male bipolar independent cohort than universal biomarkers and show evidence for a possible advantage of personalization. We identified completely novel biomarkers (such as SPTBN1 and C7orf73), and reinforced previously known biomarkers (such as PTEN and SAT1). For diagnostic ability testing purposes, we also examined as predictors phenotypic measures as apps (for suicide risk (CFI-S, Convergent Functional Information for Suicidality) and for anxiety and mood (SASS, Simplified Affective State Scale)) by themselves, as well as in combination with the top biomarkers (the combination being our a priori primary endpoint), to provide context and enhance precision of predictions. We obtained area under the curves of 90% for SI and 77% for future hospitalizations in independent cohorts. Step 5 was to look for mechanistic understanding, starting with examining evidence for the Top Dozen and Bonferroni biomarkers for involvement in other psychiatric and non-psychiatric disorders, as a mechanism for biological predisposition and vulnerability. The biomarkers we identified also provide a window towards understanding the biology of suicide, implicating biological pathways related to neurogenesis, programmed cell death and insulin signaling from the universal biomarkers, as well as mTOR signaling from the male bipolar biomarkers. In particular, HTR2A increase coupled with ARRB1 and GSK3B decreases in expression in suicidality may provide a synergistic mechanistical corrective target, as do SLC4A4 increase coupled with AHCYL1 and AHCYL2 decrease. Step 6 was to move beyond diagnostics and mechanistical risk assessment, towards providing a foundation for personalized therapeutics. Items scored positive in the CFI-S and subtypes identified by SASS in different individuals provide targets for personalized (psycho)therapy. Some individual biomarkers are targets of existing drugs used to treat mood disorders and suicidality (lithium, clozapine and omega-3 fatty acids), providing a means toward pharmacogenomics stratification of patients and monitoring of response to treatment. Such biomarkers merit evaluation in clinical trials. Bioinformatics drug repurposing analyses with the gene expression biosignatures of the Top Dozen and Bonferroni-validated universal biomarkers identified novel potential therapeutics for suicidality, such as ebselen (a lithium mimetic), piracetam (a nootropic), chlorogenic acid (a polyphenol) and metformin (an antidiabetic and possible longevity promoting drug). Finally, based on the totality of our data and of the evidence in the field to date, a convergent functional evidence score prioritizing biomarkers that have all around evidence (track suicidality, predict it, are reflective of biological predisposition and are potential drug targets) brought to the fore APOE and IL6 from among the universal biomarkers, suggesting an inflammatory/accelerated aging component that may be a targetable common denominator.
Collapse
Affiliation(s)
- A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA,Indianapolis VA Medical Center, Indianapolis, IN, USA,INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Psychiatry, Indiana University School of Medicine, Neuroscience Research Building 200B, 320 West 15th Street, Indianapolis, IN 46202, USA. E-mail:
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - P L Phalen
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H L Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Roseberry
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E M Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J O Niezer
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Williams
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D L Graham
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - T J Jones
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - V Venugopal
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Ballew
- Marion County Coroner’s Office, Indianapolis, IN, USA
| | - M Yard
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T Gelbart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Schork
- J. Craig Venter Institute, La Jolla, CA, USA
| | - G E Sandusky
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
16
|
Trifilieff P, Ducrocq F, van der Veldt S, Martinez D. Blunted Dopamine Transmission in Addiction: Potential Mechanisms and Implications for Behavior. Semin Nucl Med 2016; 47:64-74. [PMID: 27987559 DOI: 10.1053/j.semnuclmed.2016.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Positron emission tomography (PET) imaging consistently shows blunted striatal dopamine release and decreased dopamine D2 receptor availability in addiction. Here, we review the preclinical and clinical studies indicating that this neurobiological phenotype is likely to be both a consequence of chronic drug consumption and a vulnerability factor in the development of addiction. We propose that, behaviorally, blunted striatal dopamine transmission could reflect the increased impulsivity and altered cost/benefit computations that are associated with addiction. The factors that influence blunted striatal dopamine transmission in addiction are unknown. Herein, we give an overview of various factors, genetic, environmental, and social, that are known to affect dopamine transmission and that have been associated with the vulnerability to develop addiction. Altogether, these data suggest that blunted dopamine transmission and decreased D2 receptor availability are biomarkers both for the development of addiction and resistance to treatment. These findings support the view that blunted dopamine reflects impulsive behavior and deficits in motivation, which lead to the escalation of drug use.
Collapse
Affiliation(s)
- Pierre Trifilieff
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France.
| | - Fabien Ducrocq
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France
| | - Suzanne van der Veldt
- Nutrition et Neurobiologie Intégrée, INRA UMR 1286, University of Bordeaux, Bordeaux, France; Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Diana Martinez
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical College, New York, NY.
| |
Collapse
|
17
|
Rangaraju S, Levey DF, Nho K, Jain N, Andrews KD, Le-Niculescu H, Salomon DR, Saykin AJ, Petrascheck M, Niculescu AB. Mood, stress and longevity: convergence on ANK3. Mol Psychiatry 2016; 21:1037-49. [PMID: 27217151 PMCID: PMC9798616 DOI: 10.1038/mp.2016.65] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 01/01/2023]
Abstract
Antidepressants have been shown to improve longevity in C. elegans. It is plausible that orthologs of genes involved in mood regulation and stress response are involved in such an effect. We sought to understand the underlying biology. First, we analyzed the transcriptome from worms treated with the antidepressant mianserin, previously identified in a large-scale unbiased drug screen as promoting increased lifespan in worms. We identified the most robust treatment-related changes in gene expression, and identified the corresponding human orthologs. Our analysis uncovered a series of genes and biological pathways that may be at the interface between antidepressant effects and longevity, notably pathways involved in drug metabolism/degradation (nicotine and melatonin). Second, we examined which of these genes overlap with genes which may be involved in depressive symptoms in an aging non-psychiatric human population (n=3577), discovered using a genome-wide association study (GWAS) approach in a design with extremes of distribution of phenotype. Third, we used a convergent functional genomics (CFG) approach to prioritize these genes for relevance to mood disorders and stress. The top gene identified was ANK3. To validate our findings, we conducted genetic and gene-expression studies, in C. elegans and in humans. We studied C. elegans inactivating mutants for ANK3/unc-44, and show that they survive longer than wild-type, particularly in older worms, independently of mianserin treatment. We also show that some ANK3/unc-44 expression is necessary for the effects of mianserin on prolonging lifespan and survival in the face of oxidative stress, particularly in younger worms. Wild-type ANK3/unc-44 increases in expression with age in C. elegans, and is maintained at lower youthful levels by mianserin treatment. These lower levels may be optimal in terms of longevity, offering a favorable balance between sufficient oxidative stress resistance in younger worms and survival effects in older worms. Thus, ANK3/unc-44 may represent an example of antagonistic pleiotropy, in which low-expression level in young animals are beneficial, but the age-associated increase becomes detrimental. Inactivating mutations in ANK3/unc-44 reverse this effect and cause detrimental effects in young animals (sensitivity to oxidative stress) and beneficial effect in old animals (increased survival). In humans, we studied if the most significant single nucleotide polymorphism (SNP) for depressive symptoms in ANK3 from our GWAS has a relationship to lifespan, and show a trend towards longer lifespan in individuals with the risk allele for depressive symptoms in men (odds ratio (OR) 1.41, P=0.031) but not in women (OR 1.08, P=0.33). We also examined whether ANK3, by itself or in a panel with other top CFG-prioritized genes, acts as a blood gene-expression biomarker for biological age, in two independent cohorts, one of live psychiatric patients (n=737), and one of suicide completers from the coroner's office (n=45). We show significantly lower levels of ANK3 expression in chronologically younger individuals than in middle age individuals, with a diminution of that effect in suicide completers, who presumably have been exposed to more severe and acute negative mood and stress. Of note, ANK3 was previously reported to be overexpressed in fibroblasts from patients with Hutchinson-Gilford progeria syndrome, a form of accelerated aging. Taken together, these studies uncover ANK3 and other genes in our dataset as biological links between mood, stress and longevity/aging, that may be biomarkers as well as targets for preventive or therapeutic interventions. Drug repurposing bioinformatics analyses identified the relatively innocuous omega-3 fatty acid DHA (docosahexaenoic acid), piracetam, quercetin, vitamin D and resveratrol as potential longevity promoting compounds, along with a series of existing drugs, such as estrogen-like compounds, antidiabetics and sirolimus/rapamycin. Intriguingly, some of our top candidate genes for mood and stress-modulated longevity were changed in expression in opposite direction in previous studies in the Alzheimer disease. Additionally, a whole series of others were changed in expression in opposite direction in our previous studies on suicide, suggesting the possibility of a "life switch" actively controlled by mood and stress.
Collapse
Affiliation(s)
- S Rangaraju
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - DF Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Nho
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N Jain
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - KD Andrews
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - DR Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - AJ Saykin
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M Petrascheck
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - AB Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
18
|
Levey DF, Niculescu EM, Le-Niculescu H, Dainton HL, Phalen PL, Ladd TB, Weber H, Belanger E, Graham DL, Khan FN, Vanipenta NP, Stage EC, Ballew A, Yard M, Gelbart T, Shekhar A, Schork NJ, Kurian SM, Sandusky GE, Salomon DR, Niculescu AB. Towards understanding and predicting suicidality in women: biomarkers and clinical risk assessment. Mol Psychiatry 2016; 21:768-85. [PMID: 27046645 DOI: 10.1038/mp.2016.31] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/27/2016] [Accepted: 02/11/2016] [Indexed: 02/06/2023]
Abstract
Women are under-represented in research on suicidality to date. Although women have a lower rate of suicide completion than men, due in part to the less-violent methods used, they have a higher rate of suicide attempts. Our group has previously identified genomic (blood gene expression biomarkers) and clinical information (apps) predictors for suicidality in men. We now describe pilot studies in women. We used a powerful within-participant discovery approach to identify genes that change in expression between no suicidal ideation (no SI) and high suicidal ideation (high SI) states (n=12 participants out of a cohort of 51 women psychiatric participants followed longitudinally, with diagnoses of bipolar disorder, depression, schizoaffective disorder and schizophrenia). We then used a Convergent Functional Genomics (CFG) approach to prioritize the candidate biomarkers identified in the discovery step by using all the prior evidence in the field. Next, we validated for suicidal behavior the top-ranked biomarkers for SI, in a demographically matched cohort of women suicide completers from the coroner's office (n=6), by assessing which markers were stepwise changed from no SI to high SI to suicide completers. We then tested the 50 biomarkers that survived Bonferroni correction in the validation step, as well as top increased and decreased biomarkers from the discovery and prioritization steps, in a completely independent test cohort of women psychiatric disorder participants for prediction of SI (n=33) and in a future follow-up cohort of psychiatric disorder participants for prediction of psychiatric hospitalizations due to suicidality (n=24). Additionally, we examined how two clinical instruments in the form of apps, Convergent Functional Information for Suicidality (CFI-S) and Simplified Affective State Scale (SASS), previously tested in men, perform in women. The top CFI-S item distinguishing high SI from no SI states was the chronic stress of social isolation. We then showed how the clinical information apps combined with the 50 validated biomarkers into a broad predictor (UP-Suicide), our apriori primary end point, predicts suicidality in women. UP-Suicide had a receiver-operating characteristic (ROC) area under the curve (AUC) of 82% for predicting SI and an AUC of 78% for predicting future hospitalizations for suicidality. Some of the individual components of the UP-Suicide showed even better results. SASS had an AUC of 81% for predicting SI, CFI-S had an AUC of 84% and the combination of the two apps had an AUC of 87%. The top biomarker from our sequential discovery, prioritization and validation steps, BCL2, predicted future hospitalizations due to suicidality with an AUC of 89%, and the panel of 50 validated biomarkers (BioM-50) predicted future hospitalizations due to suicidality with an AUC of 94%. The best overall single blood biomarker for predictions was PIK3C3 with an AUC of 65% for SI and an AUC of 90% for future hospitalizations. Finally, we sought to understand the biology of the biomarkers. BCL2 and GSK3B, the top CFG scoring validated biomarkers, as well as PIK3C3, have anti-apoptotic and neurotrophic effects, are decreased in expression in suicidality and are known targets of the anti-suicidal mood stabilizer drug lithium, which increases their expression and/or activity. Circadian clock genes were overrepresented among the top markers. Notably, PER1, increased in expression in suicidality, had an AUC of 84% for predicting future hospitalizations, and CSNK1A1, decreased in expression, had an AUC of 96% for predicting future hospitalizations. Circadian clock abnormalities are related to mood disorder, and sleep abnormalities have been implicated in suicide. Docosahexaenoic acid signaling was one of the top biological pathways overrepresented in validated biomarkers, which is of interest given the potential therapeutic and prophylactic benefits of omega-3 fatty acids. Some of the top biomarkers from the current work in women showed co-directionality of change in expression with our previous work in men, whereas others had changes in opposite directions, underlying the issue of biological context and differences in suicidality between the two genders. With this study, we begin to shed much needed light in the area of female suicidality, identify useful objective predictors and help understand gender commonalities and differences. During the conduct of the study, one participant committed suicide. In retrospect, when the analyses were completed, her UP-Suicide risk prediction score was at the 100 percentile of all participants tested.
Collapse
Affiliation(s)
- D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E M Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H L Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - P L Phalen
- Indianapolis Veterans' Affairs Medical Center, Indianapolis, IN, USA
| | - T B Ladd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Weber
- Indianapolis Veterans' Affairs Medical Center, Indianapolis, IN, USA
| | - E Belanger
- Indianapolis Veterans' Affairs Medical Center, Indianapolis, IN, USA
| | - D L Graham
- Indianapolis Veterans' Affairs Medical Center, Indianapolis, IN, USA
| | - F N Khan
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N P Vanipenta
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E C Stage
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Ballew
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - M Yard
- Indiana Center for Biomarker Research in Neuropsychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T Gelbart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Schork
- J. Craig Venter Institute, La Jolla, CA, USA
| | - S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - G E Sandusky
- Indiana Center for Biomarker Research in Neuropsychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D R Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,Indianapolis Veterans' Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
19
|
Noda Y, Silverstein WK, Barr MS, Vila-Rodriguez F, Downar J, Rajji TK, Fitzgerald PB, Mulsant BH, Vigod SN, Daskalakis ZJ, Blumberger DM. Neurobiological mechanisms of repetitive transcranial magnetic stimulation of the dorsolateral prefrontal cortex in depression: a systematic review. Psychol Med 2015; 45:3411-3432. [PMID: 26349810 DOI: 10.1017/s0033291715001609] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Depression is one of the most prevalent mental illnesses worldwide and a leading cause of disability, especially in the setting of treatment resistance. In recent years, repetitive transcranial magnetic stimulation (rTMS) has emerged as a promising alternative strategy for treatment-resistant depression and its clinical efficacy has been investigated intensively across the world. However, the underlying neurobiological mechanisms of the antidepressant effect of rTMS are still not fully understood. This review aims to systematically synthesize the literature on the neurobiological mechanisms of treatment response to rTMS in patients with depression. Medline (1996-2014), Embase (1980-2014) and PsycINFO (1806-2014) were searched under set terms. Three authors reviewed each article and came to consensus on the inclusion and exclusion criteria. All eligible studies were reviewed, duplicates were removed, and data were extracted individually. Of 1647 articles identified, 66 studies met both inclusion and exclusion criteria. rTMS affects various biological factors that can be measured by current biological techniques. Although a number of studies have explored the neurobiological mechanisms of rTMS, a large variety of rTMS protocols and parameters limits the ability to synthesize these findings into a coherent understanding. However, a convergence of findings suggest that rTMS exerts its therapeutic effects by altering levels of various neurochemicals, electrophysiology as well as blood flow and activity in the brain in a frequency-dependent manner. More research is needed to delineate the neurobiological mechanisms of the antidepressant effect of rTMS. The incorporation of biological assessments into future rTMS clinical trials will help in this regard.
Collapse
Affiliation(s)
- Y Noda
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| | - W K Silverstein
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| | - M S Barr
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| | - F Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies Laboratory,Department of Psychiatry,Faculty of Medicine,University of British Columbia,Vancouver,British Columbia,Canada
| | - J Downar
- Department of Psychiatry,University of Toronto,Toronto,Ontario,Canada
| | - T K Rajji
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| | - P B Fitzgerald
- Monash Alfred Psychiatry Research Centre,The Alfred and Monash University Central Clinical School,Melbourne,Victoria,Australia
| | - B H Mulsant
- Department of Psychiatry,University of Toronto,Toronto,Ontario,Canada
| | - S N Vigod
- Department of Psychiatry,University of Toronto,Toronto,Ontario,Canada
| | - Z J Daskalakis
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| | - D M Blumberger
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health,Toronto,Ontario,Canada
| |
Collapse
|
20
|
Arunagiri P, Rajeshwaran K, Shanthakumar J, Tamilselvan T, Balamurugan E. Combination of omega-3 Fatty acids, lithium, and aripiprazole reduces oxidative stress in brain of mice with mania. Biol Trace Elem Res 2014; 160:409-17. [PMID: 25035188 DOI: 10.1007/s12011-014-0067-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 07/07/2014] [Indexed: 10/25/2022]
Abstract
Manic episode in bipolar disorder (BD) was evaluated in the present study with supplementation of omega-3 fatty acids in combination with aripiprazole and lithium on methylphenidate (MPD)-induced manic mice model. Administration of MPD 5 mg/kg bw intraperitoneally (i.p.) caused increase in oxidative stress in mice brain. To retract this effect, supplementation of omega-3 fatty acids 1.5 ml/kg (p.o.), aripiprazole 1.5 mg/kg bw (i.p.), and lithium 50 mg/kg bw (p.o) were given to mice. Omega-3 fatty acids alone and in combination with aripiprazole- and lithium-treated groups significantly reduced the levels of superoxide dismutase (SOD), catalase (CAT), and lipid peroxidation products (thiobarbituric acid reactive substances) in the brain. MPD treatment significantly decreased the reduced glutathione (GSH) level and glutathione peroxidase (GPx) activity, and they were restored by supplementation of omega-3 fatty acids with aripiprazole and lithium. There is no remarkable difference in the effect of creatine kinase (CK) activity between MPD-induced manic model and the treatment groups. Therefore, our results demonstrate that oxidative stress imbalance and mild insignificant CK alterations induced by administration of MPD can be restored back to normal physiological levels through omega-3 fatty acids combined with lithium and aripiprazole that attributes to effective prevention against mania in adult male Swiss albino mice.
Collapse
Affiliation(s)
- Pandiyan Arunagiri
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, 608 002, India
| | | | | | | | | |
Collapse
|
21
|
Ruby CL, Vadnie CA, Hinton DJ, Abulseoud OA, Walker DL, O'Connor KM, Noterman MF, Choi DS. Adenosinergic regulation of striatal clock gene expression and ethanol intake during constant light. Neuropsychopharmacology 2014; 39:2432-40. [PMID: 24755889 PMCID: PMC4138755 DOI: 10.1038/npp.2014.94] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/09/2014] [Accepted: 04/17/2014] [Indexed: 01/19/2023]
Abstract
Circadian rhythm and sleep disruptions occur frequently in individuals with alcohol use disorders (AUD) and present significant barriers to treatment. Recently, a variant of adenosine transporter, equilibrative nucleoside transporter 1 (ENT1), was associated with the co-occurrence of sleep problems and AUD. We have previously shown that mice lacking ENT1 (ENT1 KO) have reduced adenosine levels in the striatum and drink more alcohol compared with wild types (WT). However, it is unknown whether ENT1 deletion disrupts circadian rhythms, which may contribute to alcohol preference in ENT1 KO mice. Here we used these mice to determine whether endogenous adenosine regulates circadian genetic and behavioral rhythms and influences alcohol intake during chronodisruption. We examined circadian locomotor activity in ENT1 KO vs WT littermates and found that ENT1 KO mice were both active earlier and hyperactive compared with WT mice at night. We used real-time PCR and immunohistochemistry to estimate striatal clock gene levels and found that PER2 expression in the striatum was blunted by ENT1 deletion or A2A receptor (A2AR) antagonism. Next, we exposed ENT1 KO and WT mice to constant light (LL) and found further elevation in ethanol intake in ENT1 KO, but not in WT mice, supporting the notion that circadian dysfunction may contribute to increased alcohol intake in ENT1 KO mice. Finally, we showed that A2AR agonist administration normalized PER1 and PER2 expression and circadian locomotor activity in ENT1 KO mice. Together, our results demonstrate that adenosine signaling regulates cellular and behavioral circadian timing and influences alcohol intake during chronodisruption.
Collapse
Affiliation(s)
- Christina L Ruby
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Biology, Indiana University of Pennsylvania, Indiana, PA, USA
| | - Chelsea A Vadnie
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David J Hinton
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Denise L Walker
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katheryn M O'Connor
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria F Noterman
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA,Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA, Tel: +1 507 284 5602, Fax: +1 507 266 0824, E-mail:
| |
Collapse
|
22
|
Identification of a QTL in Mus musculus for alcohol preference, withdrawal, and Ap3m2 expression using integrative functional genomics and precision genetics. Genetics 2014; 197:1377-93. [PMID: 24923803 DOI: 10.1534/genetics.114.166165] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Extensive genetic and genomic studies of the relationship between alcohol drinking preference and withdrawal severity have been performed using animal models. Data from multiple such publications and public data resources have been incorporated in the GeneWeaver database with >60,000 gene sets including 285 alcohol withdrawal and preference-related gene sets. Among these are evidence for positional candidates regulating these behaviors in overlapping quantitative trait loci (QTL) mapped in distinct mouse populations. Combinatorial integration of functional genomics experimental results revealed a single QTL positional candidate gene in one of the loci common to both preference and withdrawal. Functional validation studies in Ap3m2 knockout mice confirmed these relationships. Genetic validation involves confirming the existence of segregating polymorphisms that could account for the phenotypic effect. By exploiting recent advances in mouse genotyping, sequence, epigenetics, and phylogeny resources, we confirmed that Ap3m2 resides in an appropriately segregating genomic region. We have demonstrated genetic and alcohol-induced regulation of Ap3m2 expression. Although sequence analysis revealed no polymorphisms in the Ap3m2-coding region that could account for all phenotypic differences, there are several upstream SNPs that could. We have identified one of these to be an H3K4me3 site that exhibits strain differences in methylation. Thus, by making cross-species functional genomics readily computable we identified a common QTL candidate for two related bio-behavioral processes via functional evidence and demonstrate sufficiency of the genetic locus as a source of variation underlying two traits.
Collapse
|
23
|
Genetic risk prediction and neurobiological understanding of alcoholism. Transl Psychiatry 2014; 4:e391. [PMID: 24844177 PMCID: PMC4035721 DOI: 10.1038/tp.2014.29] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/18/2014] [Indexed: 01/08/2023] Open
Abstract
We have used a translational Convergent Functional Genomics (CFG) approach to discover genes involved in alcoholism, by gene-level integration of genome-wide association study (GWAS) data from a German alcohol dependence cohort with other genetic and gene expression data, from human and animal model studies, similar to our previous work in bipolar disorder and schizophrenia. A panel of all the nominally significant P-value SNPs in the top candidate genes discovered by CFG (n=135 genes, 713 SNPs) was used to generate a genetic risk prediction score (GRPS), which showed a trend towards significance (P=0.053) in separating alcohol dependent individuals from controls in an independent German test cohort. We then validated and prioritized our top findings from this discovery work, and subsequently tested them in three independent cohorts, from two continents. A panel of all the nominally significant P-value single-nucleotide length polymorphisms (SNPs) in the top candidate genes discovered by CFG (n=135 genes, 713 SNPs) were used to generate a Genetic Risk Prediction Score (GRPS), which showed a trend towards significance (P=0.053) in separating alcohol-dependent individuals from controls in an independent German test cohort. In order to validate and prioritize the key genes that drive behavior without some of the pleiotropic environmental confounds present in humans, we used a stress-reactive animal model of alcoholism developed by our group, the D-box binding protein (DBP) knockout mouse, consistent with the surfeit of stress theory of addiction proposed by Koob and colleagues. A much smaller panel (n=11 genes, 66 SNPs) of the top CFG-discovered genes for alcoholism, cross-validated and prioritized by this stress-reactive animal model showed better predictive ability in the independent German test cohort (P=0.041). The top CFG scoring gene for alcoholism from the initial discovery step, synuclein alpha (SNCA) remained the top gene after the stress-reactive animal model cross-validation. We also tested this small panel of genes in two other independent test cohorts from the United States, one with alcohol dependence (P=0.00012) and one with alcohol abuse (a less severe form of alcoholism; P=0.0094). SNCA by itself was able to separate alcoholics from controls in the alcohol-dependent cohort (P=0.000013) and the alcohol abuse cohort (P=0.023). So did eight other genes from the panel of 11 genes taken individually, albeit to a lesser extent and/or less broadly across cohorts. SNCA, GRM3 and MBP survived strict Bonferroni correction for multiple comparisons. Taken together, these results suggest that our stress-reactive DBP animal model helped to validate and prioritize from the CFG-discovered genes some of the key behaviorally relevant genes for alcoholism. These genes fall into a series of biological pathways involved in signal transduction, transmission of nerve impulse (including myelination) and cocaine addiction. Overall, our work provides leads towards a better understanding of illness, diagnostics and therapeutics, including treatment with omega-3 fatty acids. We also examined the overlap between the top candidate genes for alcoholism from this work and the top candidate genes for bipolar disorder, schizophrenia, anxiety from previous CFG analyses conducted by us, as well as cross-tested genetic risk predictions. This revealed the significant genetic overlap with other major psychiatric disorder domains, providing a basis for comorbidity and dual diagnosis, and placing alcohol use in the broader context of modulating the mental landscape.
Collapse
|
24
|
Hammamieh R, Chakraborty N, Gautam A, Miller SA, Muhie S, Meyerhoff J, Jett M. Transcriptomic analysis of the effects of a fish oil enriched diet on murine brains. PLoS One 2014; 9:e90425. [PMID: 24632812 PMCID: PMC3954562 DOI: 10.1371/journal.pone.0090425] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/29/2014] [Indexed: 12/15/2022] Open
Abstract
The health benefits of fish oil enriched with high omega-3 polyunsaturated fatty acids (n-3 PUFA) are widely documented. Fish oil as dietary supplements, however, show moderate clinical efficacy, highlighting an immediate scope of systematic in vitro feedback. Our transcriptomic study was designed to investigate the genomic shift of murine brains fed on fish oil enriched diets. A customized fish oil enriched diet (FD) and standard lab diet (SD) were separately administered to two randomly chosen populations of C57BL/6J mice from their weaning age until late adolescence. Statistical analysis mined 1,142 genes of interest (GOI) differentially altered in the hemibrains collected from the FD- and SD-fed mice at the age of five months. The majority of identified GOI (∼40%) encodes proteins located in the plasma membrane, suggesting that fish oil primarily facilitated the membrane-oriented biofunctions. FD potentially augmented the nervous system's development and functions by selectively stimulating the Src-mediated calcium-induced growth cascade and the downstream PI3K-AKT-PKC pathways. FD reduced the amyloidal burden, attenuated oxidative stress, and assisted in somatostatin activation—the signatures of attenuation of Alzheimer's disease, Parkinson's disease, and affective disorder. FD induced elevation of FKBP5 and suppression of BDNF, which are often linked with the improvement of anxiety disorder, depression, and post-traumatic stress disorder. Hence we anticipate efficacy of FD in treating illnesses such as depression that are typically triggered by the hypoactivities of dopaminergic, adrenergic, cholinergic, and GABAergic networks. Contrastingly, FD's efficacy could be compromised in treating illnesses such as bipolar disorder and schizophrenia, which are triggered by hyperactivities of the same set of neuromodulators. A more comprehensive investigation is recommended to elucidate the implications of fish oil on disease pathomechanisms, and the result-driven repositioning of fish oil utilization may revitalize its therapeutic efficacy.
Collapse
Affiliation(s)
- Rasha Hammamieh
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
- * E-mail:
| | - Nabarun Chakraborty
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| | - Aarti Gautam
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| | - Stacy-Ann Miller
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| | - Seid Muhie
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| | - James Meyerhoff
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| | - Marti Jett
- United States Army Center for Environmental Health Research, Fort Detrick, Maryland, United States of America
| |
Collapse
|
25
|
Discovery and validation of blood biomarkers for suicidality. Mol Psychiatry 2013; 18:1249-64. [PMID: 23958961 PMCID: PMC3835939 DOI: 10.1038/mp.2013.95] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/21/2013] [Accepted: 06/25/2013] [Indexed: 01/01/2023]
Abstract
Suicides are a leading cause of death in psychiatric patients, and in society at large. Developing more quantitative and objective ways (biomarkers) for predicting and tracking suicidal states would have immediate practical applications and positive societal implications. We undertook such an endeavor. First, building on our previous blood biomarker work in mood disorders and psychosis, we decided to identify blood gene expression biomarkers for suicidality, looking at differential expression of genes in the blood of subjects with a major mood disorder (bipolar disorder), a high-risk population prone to suicidality. We compared no suicidal ideation (SI) states and high SI states using a powerful intrasubject design, as well as an intersubject case-case design, to generate a list of differentially expressed genes. Second, we used a comprehensive Convergent Functional Genomics (CFG) approach to identify and prioritize from the list of differentially expressed gene biomarkers of relevance to suicidality. CFG integrates multiple independent lines of evidence-genetic and functional genomic data-as a Bayesian strategy for identifying and prioritizing findings, reducing the false-positives and false-negatives inherent in each individual approach. Third, we examined whether expression levels of the blood biomarkers identified by us in the live bipolar subject cohort are actually altered in the blood in an age-matched cohort of suicide completers collected from the coroner's office, and report that 13 out of the 41 top CFG scoring biomarkers (32%) show step-wise significant change from no SI to high SI states, and then to the suicide completers group. Six out of them (15%) remained significant after strict Bonferroni correction for multiple comparisons. Fourth, we show that the blood levels of SAT1 (spermidine/spermine N1-acetyltransferase 1), the top biomarker identified by us, at the time of testing for this study, differentiated future as well as past hospitalizations with suicidality, in a live cohort of bipolar disorder subjects, and exhibited a similar but weaker pattern in a live cohort of psychosis (schizophrenia/schizoaffective disorder) subjects. Three other (phosphatase and tensin homolog (PTEN), myristoylated alanine-rich protein kinase C substrate (MARCKS), and mitogen-activated protein kinase kinase kinase 3 (MAP3K3)) of the six biomarkers that survived Bonferroni correction showed similar but weaker effects. Taken together, the prospective and retrospective hospitalization data suggests SAT1, PTEN, MARCKS and MAP3K3 might be not only state biomarkers but trait biomarkers as well. Fifth, we show how a multi-dimensional approach using SAT1 blood expression levels and two simple visual-analog scales for anxiety and mood enhances predictions of future hospitalizations for suicidality in the bipolar cohort (receiver-operating characteristic curve with area under the curve of 0.813). Of note, this simple approach does not directly ask about SI, which some individuals may deny or choose not to share with clinicians. Lastly, we conducted bioinformatic analyses to identify biological pathways, mechanisms and medication targets. Overall, suicidality may be underlined, at least in part, by biological mechanisms related to stress, inflammation and apoptosis.
Collapse
|
26
|
Niculescu AB. Convergent functional genomics of psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:587-94. [PMID: 23728881 DOI: 10.1002/ajmg.b.32163] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/19/2013] [Indexed: 12/27/2022]
Abstract
Genetic and gene expression studies, in humans and animal models of psychiatric and other medical disorders, are becoming increasingly integrated. Particularly for genomics, the convergence and integration of data across species, experimental modalities and technical platforms is providing a fit-to-disease way of extracting reproducible and biologically important signal, in contrast to the fit-to-cohort effect and limited reproducibility of human genetic analyses alone. With the advent of whole-genome sequencing and the realization that a major portion of the non-coding genome may contain regulatory variants, Convergent Functional Genomics (CFG) approaches are going to be essential to identify disease-relevant signal from the tremendous polymorphic variation present in the general population. Such work in psychiatry can provide an example of how to address other genetically complex disorders, and in turn will benefit by incorporating concepts from other areas, such as cancer, cardiovascular diseases, and diabetes.
Collapse
Affiliation(s)
- Alexander B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana; Indianapolis VA Medical Center, Indianapolis, Indiana
| |
Collapse
|
27
|
Abstract
While antidepressant therapy is an essential treatment of major depression, a substantial group of treated patients do not respond to therapy, or suffer from severe side effects. Moreover, the time of onset of the clinical improvement is often delayed. Antidepressants as currently available usually enhance serotonergic, noradrenergic and dopaminergic neurotransmission and may contribute to the inadequate remission rates for major depression. Therefore biomarkers enabling the identification of subgroups of patients and also finding unprecedented targets would provide the basis for personalized medication and thus improve treatment efficacy and reduce side effects. Several pharmacogenetic studies on antidepressant treatment response using single nucleotide polymorphism (SNPs) mapping have been performed but provided only modest findings. Therefore the analysis of gene expression to integrate genomic activity and environmental effects promises a new approach to cope with the complexity of factors influencing antidepressant treatment. Here gene expression studies focusing on candidate genes and genome-wide approaches using RNA derived from peripheral blood cells are reviewed. The most promising findings exist for hypothalamic-pituitary-adrenal (HPA) axis, inflammation and neuroplasticity related genes. However, straightforward translation into tailored treatment is still unlikely. Contradictory results limit the clinical use of the findings. Future studies are necessary, which could include functional analysis and consider gene-environment interactions.
Collapse
Affiliation(s)
- Andreas Menke
- Max Planck Institute of Psychiatry , Munich , Germany
| |
Collapse
|
28
|
Camacho A. Is anxious-depression an inflammatory state? Med Hypotheses 2013; 81:577-81. [PMID: 23891039 PMCID: PMC3782383 DOI: 10.1016/j.mehy.2013.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/24/2013] [Accepted: 07/03/2013] [Indexed: 12/22/2022]
Abstract
For several years, the literature has examined the association of depression and anxiety with inflammatory states such as atherosclerosis and cardiovascular disease, yet this association remains inconclusive. Several possible immune and endocrinological pathways have been postulated that associate depression and anxiety with inflammation and immune dysregulation. Anxiety and depression have usually been envisioned as two separate psychiatric conditions yet they share similar symptoms and are frequently encountered together among individuals. Individuals suffering from anxious-depression are more refractory to treatment and have been reported to have greater disability compared to individuals with anxiety or depression alone. With the current changes in the diagnostic manual for psychiatric disorders placing more emphasis on a dimensional approach for the diagnosis of psychiatric illnesses, the hypothesis presented is that anxious-depression should be considered as a chronic inflammatory phenomenon since it shares common physiopathological pathways and pharmacological treatments with inflammatory states. This hypothesis might help to investigate how different levels of inflammatory biomarkers could be correlated with symptoms of anxious-depression.
Collapse
Affiliation(s)
- Alvaro Camacho
- Department of Family and Preventive Medicine, University of California, San Diego, United States; Department of Psychiatry, University of California, San Diego, United States.
| |
Collapse
|
29
|
Abstract
Stem cell technologies provide an exciting avenue to directly access the transcriptome of patients in neuronal-like cell types, which might have more direct relevance to brain research than other peripheral tissues (blood, fibroblasts). Enthusiasm should be tempered by concerns that artifacts and noise might be generated as part of the in vitro process of creating and maintaining these cell type. A solution may be to apply a Convergent Functional Genomics approach, where the data from stem cell-derived neuronal cells are integrated, cross-validated and prioritized using independent lines of evidence from other approaches and platforms (human genetic data, human postmortem brain data, animal model data). I provide a brief overview and an example in support of such an approach.
Collapse
|
30
|
Low frequency transcranial magnetic stimulation downregulates expression of stress genes in blood leucocytes: preliminary evidence. J Psychiatr Res 2013; 47:935-6. [PMID: 23548330 DOI: 10.1016/j.jpsychires.2013.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 02/14/2013] [Accepted: 03/05/2013] [Indexed: 01/07/2023]
|
31
|
Teyssier JR, Chauvet-Gelinier JC, Ragot S, Bonin B. Up-regulation of leucocytes genes implicated in telomere dysfunction and cellular senescence correlates with depression and anxiety severity scores. PLoS One 2012. [PMID: 23185405 PMCID: PMC3504145 DOI: 10.1371/journal.pone.0049677] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Major depressive disorder (MDD) is frequently associated with chronic medical illness responsible of increased disability and mortality. Inflammation and oxidative stress are considered to be the major mediators of the allostatic load, and has been shown to correlate with telomere erosion in the leucocytes of MDD patients, leading to the model of accelerated aging. However, the significance of telomere length as an exclusive biomarker of aging has been questioned on both methodological and biological grounds. Furthermore, telomeres significantly shorten only in patients with long lasting MDD. Sensitive and dynamic functional biomarkers of aging would be clinically useful to evaluate the somatic impact of MDD. Methodology To address this issue we have measured in the blood leucocytes of MDD patients (N = 17) and controls (N = 16) the expression of two genes identified as robust biomarkers of human aging and telomere dysfunction: p16INK4a and STMN1. We have also quantified the transcripts of genes involved in the repair of oxidative DNA damage at telomeres (OGG1), telomere regulation and elongation (TERT), and in the response to biopsychological stress (FOS and DUSP1). Results The OGG1, p16INK4a, and STMN1 gene were significantly up-regulated (25 to 100%) in the leucocytes of MDD patients. Expression of p16INK4a and STMN1 was directly correlated with anxiety scores in the depression group, and that of p16INK4a, STMN and TERT with the depression and anxiety scores in the combined sample (MDD plus controls). Furthermore, we identified a unique correlative pattern of gene expression in the leucocytes of MDD subjects. Conclusions Expression of p16INK4 and STMN1 is a promising biomarker for future epidemiological assessment of the somatic impact of depressive and anxious symptoms, at both clinical and subclinical level in both depressive patients and general population.
Collapse
Affiliation(s)
- Jean-Raymond Teyssier
- Department of Genetics and Laboratory of Molecular Genetics, University Hospital, Dijon, France.
| | | | | | | |
Collapse
|
32
|
Ayalew M, Le-Niculescu H, Levey DF, Jain N, Changala B, Patel SD, Winiger E, Breier A, Shekhar A, Amdur R, Koller D, Nurnberger JI, Corvin A, Geyer M, Tsuang MT, Salomon D, Schork NJ, Fanous AH, O'Donovan MC, Niculescu AB. Convergent functional genomics of schizophrenia: from comprehensive understanding to genetic risk prediction. Mol Psychiatry 2012; 17:887-905. [PMID: 22584867 PMCID: PMC3427857 DOI: 10.1038/mp.2012.37] [Citation(s) in RCA: 311] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/28/2012] [Accepted: 03/05/2012] [Indexed: 02/07/2023]
Abstract
We have used a translational convergent functional genomics (CFG) approach to identify and prioritize genes involved in schizophrenia, by gene-level integration of genome-wide association study data with other genetic and gene expression studies in humans and animal models. Using this polyevidence scoring and pathway analyses, we identify top genes (DISC1, TCF4, MBP, MOBP, NCAM1, NRCAM, NDUFV2, RAB18, as well as ADCYAP1, BDNF, CNR1, COMT, DRD2, DTNBP1, GAD1, GRIA1, GRIN2B, HTR2A, NRG1, RELN, SNAP-25, TNIK), brain development, myelination, cell adhesion, glutamate receptor signaling, G-protein-coupled receptor signaling and cAMP-mediated signaling as key to pathophysiology and as targets for therapeutic intervention. Overall, the data are consistent with a model of disrupted connectivity in schizophrenia, resulting from the effects of neurodevelopmental environmental stress on a background of genetic vulnerability. In addition, we show how the top candidate genes identified by CFG can be used to generate a genetic risk prediction score (GRPS) to aid schizophrenia diagnostics, with predictive ability in independent cohorts. The GRPS also differentiates classic age of onset schizophrenia from early onset and late-onset disease. We also show, in three independent cohorts, two European American and one African American, increasing overlap, reproducibility and consistency of findings from single-nucleotide polymorphisms to genes, then genes prioritized by CFG, and ultimately at the level of biological pathways and mechanisms. Finally, we compared our top candidate genes for schizophrenia from this analysis with top candidate genes for bipolar disorder and anxiety disorders from previous CFG analyses conducted by us, as well as findings from the fields of autism and Alzheimer. Overall, our work maps the genomic and biological landscape for schizophrenia, providing leads towards a better understanding of illness, diagnostics and therapeutics. It also reveals the significant genetic overlap with other major psychiatric disorder domains, suggesting the need for improved nosology.
Collapse
Affiliation(s)
- M Ayalew
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - D F Levey
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N Jain
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - B Changala
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Winiger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Breier
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Amdur
- Washington DC VA Medical Center, Washington, DC, USA
| | - D Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J I Nurnberger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Corvin
- Department of Psychiatry, Trinity College, Dublin, Ireland
| | - M Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - M T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - D Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - N J Schork
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - A H Fanous
- Washington DC VA Medical Center, Washington, DC, USA
| | - M C O'Donovan
- Department of Psychological Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
33
|
Stelzer G, Dalah I, Stein TI, Satanower Y, Rosen N, Nativ N, Oz-Levi D, Olender T, Belinky F, Bahir I, Krug H, Perco P, Mayer B, Kolker E, Safran M, Lancet D. In-silico human genomics with GeneCards. Hum Genomics 2012; 5:709-17. [PMID: 22155609 PMCID: PMC3525253 DOI: 10.1186/1479-7364-5-6-709] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Since 1998, the bioinformatics, systems biology, genomics and medical communities have enjoyed a synergistic relationship with the GeneCards database of human genes (http://www.genecards.org). This human gene compendium was created to help to introduce order into the increasing chaos of information flow. As a consequence of viewing details and deep links related to specific genes, users have often requested enhanced capabilities, such that, over time, GeneCards has blossomed into a suite of tools (including GeneDecks, GeneALaCart, GeneLoc, GeneNote and GeneAnnot) for a variety of analyses of both single human genes and sets thereof. In this paper, we focus on inhouse and external research activities which have been enabled, enhanced, complemented and, in some cases, motivated by GeneCards. In turn, such interactions have often inspired and propelled improvements in GeneCards. We describe here the evolution and architecture of this project, including examples of synergistic applications in diverse areas such as synthetic lethality in cancer, the annotation of genetic variations in disease, omics integration in a systems biology approach to kidney disease, and bioinformatics tools.
Collapse
Affiliation(s)
- Gil Stelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|