1
|
AudioChip: A Deep Phenotyping Approach for Deconstructing and Quantifying Audiological Phenotypes of Self-Reported Speech Perception Difficulties. Ear Hear 2021; 43:1023-1036. [PMID: 34860719 PMCID: PMC9010350 DOI: 10.1097/aud.0000000000001158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES About 15% of U.S. adults report speech perception difficulties despite showing normal audiograms. Recent research suggests that genetic factors might influence the phenotypic spectrum of speech perception difficulties. The primary objective of the present study was to describe a conceptual framework of a deep phenotyping method, referred to as AudioChipping, for deconstructing and quantifying complex audiometric phenotypes. DESIGN In a sample of 70 females 18 to 35 years of age with normal audiograms (from 250 to 8000 Hz), the study measured behavioral hearing thresholds (250 to 16,000 Hz), distortion product otoacoustic emissions (1000 to 16,000 Hz), click-evoked auditory brainstem responses (ABR), complex ABR (cABR), QuickSIN, dichotic digit test score, loudness discomfort level, and noise exposure background. The speech perception difficulties were evaluated using the Speech, Spatial, and Quality of Hearing Scale-12-item version (SSQ). A multiple linear regression model was used to determine the relationship between SSQ scores and audiometric measures. Participants were categorized into three groups (i.e., high, mid, and low) using the SSQ scores before performing the clustering analysis. Audiometric measures were normalized and standardized before performing unsupervised k-means clustering to generate AudioChip. RESULTS The results showed that SSQ and noise exposure background exhibited a significant negative correlation. ABR wave I amplitude, cABR offset latency, cABR response morphology, and loudness discomfort level were significant predictors for SSQ scores. These predictors explained about 18% of the variance in the SSQ score. The k-means clustering was used to split the participants into three major groups; one of these clusters revealed 53% of participants with low SSQ. CONCLUSIONS Our study highlighted the relationship between SSQ and auditory coding precision in the auditory brainstem in normal-hearing young females. AudioChip was useful in delineating and quantifying internal homogeneity and heterogeneity in audiometric measures among individuals with a range of SSQ scores. AudioChip could help identify the genotype-phenotype relationship, document longitudinal changes in auditory phenotypes, and pair individuals in case-control groups for the genetic association analysis.
Collapse
|
2
|
Le-Niculescu H, Roseberry K, Gill SS, Levey DF, Phalen PL, Mullen J, Williams A, Bhairo S, Voegtline T, Davis H, Shekhar A, Kurian SM, Niculescu AB. Precision medicine for mood disorders: objective assessment, risk prediction, pharmacogenomics, and repurposed drugs. Mol Psychiatry 2021; 26:2776-2804. [PMID: 33828235 PMCID: PMC8505261 DOI: 10.1038/s41380-021-01061-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/08/2021] [Accepted: 02/24/2021] [Indexed: 12/23/2022]
Abstract
Mood disorders (depression, bipolar disorders) are prevalent and disabling. They are also highly co-morbid with other psychiatric disorders. Currently there are no objective measures, such as blood tests, used in clinical practice, and available treatments do not work in everybody. The development of blood tests, as well as matching of patients with existing and new treatments, in a precise, personalized and preventive fashion, would make a significant difference at an individual and societal level. Early pilot studies by us to discover blood biomarkers for mood state were promising [1], and validated by others [2]. Recent work by us has identified blood gene expression biomarkers that track suicidality, a tragic behavioral outcome of mood disorders, using powerful longitudinal within-subject designs, validated them in suicide completers, and tested them in independent cohorts for ability to assess state (suicidal ideation), and ability to predict trait (future hospitalizations for suicidality) [3-6]. These studies showed good reproducibility with subsequent independent genetic studies [7]. More recently, we have conducted such studies also for pain [8], for stress disorders [9], and for memory/Alzheimer's Disease [10]. We endeavored to use a similar comprehensive approach to identify more definitive biomarkers for mood disorders, that are transdiagnostic, by studying mood in psychiatric disorders patients. First, we used a longitudinal within-subject design and whole-genome gene expression approach to discover biomarkers which track mood state in subjects who had diametric changes in mood state from low to high, from visit to visit, as measured by a simple visual analog scale that we had previously developed (SMS-7). Second, we prioritized these biomarkers using a convergent functional genomics (CFG) approach encompassing in a comprehensive fashion prior published evidence in the field. Third, we validated the biomarkers in an independent cohort of subjects with clinically severe depression (as measured by Hamilton Depression Scale, (HAMD)) and with clinically severe mania (as measured by the Young Mania Rating Scale (YMRS)). Adding the scores from the first three steps into an overall convergent functional evidence (CFE) score, we ended up with 26 top candidate blood gene expression biomarkers that had a CFE score as good as or better than SLC6A4, an empirical finding which we used as a de facto positive control and cutoff. Notably, there was among them an enrichment in genes involved in circadian mechanisms. We further analyzed the biological pathways and networks for the top candidate biomarkers, showing that circadian, neurotrophic, and cell differentiation functions are involved, along with serotonergic and glutamatergic signaling, supporting a view of mood as reflecting energy, activity and growth. Fourth, we tested in independent cohorts of psychiatric patients the ability of each of these 26 top candidate biomarkers to assess state (mood (SMS-7), depression (HAMD), mania (YMRS)), and to predict clinical course (future hospitalizations for depression, future hospitalizations for mania). We conducted our analyses across all patients, as well as personalized by gender and diagnosis, showing increased accuracy with the personalized approach, particularly in women. Again, using SLC6A4 as the cutoff, twelve top biomarkers had the strongest overall evidence for tracking and predicting depression after all four steps: NRG1, DOCK10, GLS, PRPS1, TMEM161B, GLO1, FANCF, HNRNPDL, CD47, OLFM1, SMAD7, and SLC6A4. Of them, six had the strongest overall evidence for tracking and predicting both depression and mania, hence bipolar mood disorders. There were also two biomarkers (RLP3 and SLC6A4) with the strongest overall evidence for mania. These panels of biomarkers have practical implications for distinguishing between depression and bipolar disorder. Next, we evaluated the evidence for our top biomarkers being targets of existing psychiatric drugs, which permits matching patients to medications in a targeted fashion, and the measuring of response to treatment. We also used the biomarker signatures to bioinformatically identify new/repurposed candidate drugs. Top drugs of interest as potential new antidepressants were pindolol, ciprofibrate, pioglitazone and adiphenine, as well as the natural compounds asiaticoside and chlorogenic acid. The last 3 had also been identified by our previous suicidality studies. Finally, we provide an example of how a report to doctors would look for a patient with depression, based on the panel of top biomarkers (12 for depression and bipolar, one for mania), with an objective depression score, risk for future depression, and risk for bipolar switching, as well as personalized lists of targeted prioritized existing psychiatric medications and new potential medications. Overall, our studies provide objective assessments, targeted therapeutics, and monitoring of response to treatment, that enable precision medicine for mood disorders.
Collapse
Affiliation(s)
- H. Le-Niculescu
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.257413.60000 0001 2287 3919Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN USA
| | - K. Roseberry
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA
| | - S. S. Gill
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA
| | - D. F. Levey
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.47100.320000000419368710Present Address: Department of Psychiatry, Yale University School of Medicine, New Haven, CT USA
| | - P. L. Phalen
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.411024.20000 0001 2175 4264Present Address: VA Maryland Health Care System/University of Maryland School of Medicine, Baltimore, MD USA
| | - J. Mullen
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA
| | - A. Williams
- grid.280828.80000 0000 9681 3540Indianapolis VA Medical Center, Indianapolis, IN USA
| | - S. Bhairo
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.280828.80000 0000 9681 3540Indianapolis VA Medical Center, Indianapolis, IN USA
| | - T. Voegtline
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.280828.80000 0000 9681 3540Indianapolis VA Medical Center, Indianapolis, IN USA
| | - H. Davis
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.280828.80000 0000 9681 3540Indianapolis VA Medical Center, Indianapolis, IN USA
| | - A. Shekhar
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.21925.3d0000 0004 1936 9000Present Address: Office of the Dean, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - S. M. Kurian
- grid.214007.00000000122199231Scripps Health and Department of Molecular Medicine, Scripps Research, La Jolla, CA USA
| | - A. B. Niculescu
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN USA ,grid.257413.60000 0001 2287 3919Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN USA ,grid.280828.80000 0000 9681 3540Indianapolis VA Medical Center, Indianapolis, IN USA
| |
Collapse
|
3
|
The protocadherin 17 gene affects cognition, personality, amygdala structure and function, synapse development and risk of major mood disorders. Mol Psychiatry 2018; 23:400-412. [PMID: 28070120 PMCID: PMC5794872 DOI: 10.1038/mp.2016.231] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/27/2016] [Accepted: 11/01/2016] [Indexed: 01/13/2023]
Abstract
Major mood disorders, which primarily include bipolar disorder and major depressive disorder, are the leading cause of disability worldwide and pose a major challenge in identifying robust risk genes. Here, we present data from independent large-scale clinical data sets (including 29 557 cases and 32 056 controls) revealing brain expressed protocadherin 17 (PCDH17) as a susceptibility gene for major mood disorders. Single-nucleotide polymorphisms (SNPs) spanning the PCDH17 region are significantly associated with major mood disorders; subjects carrying the risk allele showed impaired cognitive abilities, increased vulnerable personality features, decreased amygdala volume and altered amygdala function as compared with non-carriers. The risk allele predicted higher transcriptional levels of PCDH17 mRNA in postmortem brain samples, which is consistent with increased gene expression in patients with bipolar disorder compared with healthy subjects. Further, overexpression of PCDH17 in primary cortical neurons revealed significantly decreased spine density and abnormal dendritic morphology compared with control groups, which again is consistent with the clinical observations of reduced numbers of dendritic spines in the brains of patients with major mood disorders. Given that synaptic spines are dynamic structures which regulate neuronal plasticity and have crucial roles in myriad brain functions, this study reveals a potential underlying biological mechanism of a novel risk gene for major mood disorders involved in synaptic function and related intermediate phenotypes.
Collapse
|
4
|
Schizophrenia: A review of potential biomarkers. J Psychiatr Res 2017; 93:37-49. [PMID: 28578207 DOI: 10.1016/j.jpsychires.2017.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Understanding the biological process and progression of schizophrenia is the first step to developing novel approaches and new interventions. Research on new biomarkers is extremely important when the goal is an early diagnosis (prediction) and precise theranostics. The objective of this review is to understand the research on biomarkers and their effects in schizophrenia to synthesize the role of these new advances. METHODS In this review, we search and review publications in databases in accordance with established limits and specific objectives. We look at particular endpoints such as the category of biomarkers, laboratory techniques and the results/conclusions of the selected publications. RESULTS The investigation of biomarkers and their potential as a predictor, diagnosis instrument and therapeutic orientation, requires an appropriate methodological strategy. In this review, we found different laboratory techniques to identify biomarkers and their function in schizophrenia. CONCLUSION The consolidation of this information will provide a large-scale application network of schizophrenia biomarkers.
Collapse
|
5
|
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: 67] [Impact Index Per Article: 9.6] [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
|
6
|
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: 49] [Impact Index Per Article: 6.1] [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
|
7
|
|
8
|
Niculescu AB, Levey DF, Phalen PL, Le-Niculescu H, Dainton HD, Jain N, Belanger E, James A, George S, Weber H, Graham DL, Schweitzer R, Ladd TB, Learman R, Niculescu EM, Vanipenta NP, Khan FN, Mullen J, Shankar G, Cook S, Humbert C, Ballew A, Yard M, Gelbart T, Shekhar A, Schork NJ, Kurian SM, Sandusky GE, Salomon DR. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry 2015; 20:1266-85. [PMID: 26283638 PMCID: PMC4759104 DOI: 10.1038/mp.2015.112] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 12/26/2022]
Abstract
Worldwide, one person dies every 40 seconds by suicide, a potentially preventable tragedy. A limiting step in our ability to intervene is the lack of objective, reliable predictors. We have previously provided proof of principle for the use of blood gene expression biomarkers to predict future hospitalizations due to suicidality, in male bipolar disorder participants. We now generalize the discovery, prioritization, validation, and testing of such markers across major psychiatric disorders (bipolar disorder, major depressive disorder, schizoaffective disorder, and schizophrenia) in male participants, to understand commonalities and differences. We used a powerful within-participant discovery approach to identify genes that change in expression between no suicidal ideation and high suicidal ideation states (n=37 participants out of a cohort of 217 psychiatric participants followed longitudinally). We then used a convergent functional genomics (CFG) approach with existing prior evidence in the field to prioritize the candidate biomarkers identified in the discovery step. Next, we validated the top biomarkers from the prioritization step for relevance to suicidal behavior, in a demographically matched cohort of suicide completers from the coroner's office (n=26). The biomarkers for suicidal ideation only are enriched for genes involved in neuronal connectivity and schizophrenia, the biomarkers also validated for suicidal behavior are enriched for genes involved in neuronal activity and mood. The 76 biomarkers that survived Bonferroni correction after validation for suicidal behavior map to biological pathways involved in immune and inflammatory response, mTOR signaling and growth factor regulation. mTOR signaling is necessary for the effects of the rapid-acting antidepressant agent ketamine, providing a novel biological rationale for its possible use in treating acute suicidality. Similarly, MAOB, a target of antidepressant inhibitors, was one of the increased biomarkers for suicidality. We also identified other potential therapeutic targets or biomarkers for drugs known to mitigate suicidality, such as omega-3 fatty acids, lithium and clozapine. Overall, 14% of the top candidate biomarkers also had evidence for involvement in psychological stress response, and 19% for involvement in programmed cell death/cellular suicide (apoptosis). It may be that in the face of adversity (stress), death mechanisms are turned on at a cellular (apoptosis) and organismal level. Finally, we tested the top increased and decreased biomarkers from the discovery for suicidal ideation (CADM1, CLIP4, DTNA, KIF2C), prioritization with CFG for prior evidence (SAT1, SKA2, SLC4A4), and validation for behavior in suicide completers (IL6, MBP, JUN, KLHDC3) steps in a completely independent test cohort of psychiatric participants for prediction of suicidal ideation (n=108), and in a future follow-up cohort of psychiatric participants (n=157) for prediction of psychiatric hospitalizations due to suicidality. The best individual biomarker across psychiatric diagnoses for predicting suicidal ideation was SLC4A4, with a receiver operating characteristic (ROC) area under the curve (AUC) of 72%. For bipolar disorder in particular, SLC4A4 predicted suicidal ideation with an AUC of 93%, and future hospitalizations with an AUC of 70%. SLC4A4 is involved in brain extracellular space pH regulation. Brain pH has been implicated in the pathophysiology of acute panic attacks. We also describe two new clinical information apps, one for affective state (simplified affective state scale, SASS) and one for suicide risk factors (Convergent Functional Information for Suicide, CFI-S), and how well they predict suicidal ideation across psychiatric diagnoses (AUC of 85% for SASS, AUC of 89% for CFI-S). We hypothesized a priori, based on our previous work, that the integration of the top biomarkers and the clinical information into a universal predictive measure (UP-Suicide) would show broad-spectrum predictive ability across psychiatric diagnoses. Indeed, the UP-Suicide was able to predict suicidal ideation across psychiatric diagnoses with an AUC of 92%. For bipolar disorder, it predicted suicidal ideation with an AUC of 98%, and future hospitalizations with an AUC of 94%. Of note, both types of tests we developed (blood biomarkers and clinical information apps) do not require asking the individual assessed if they have thoughts of suicide, as individuals who are truly suicidal often do not share that information with clinicians. We propose that the widespread use of such risk prediction tests as part of routine or targeted healthcare assessments will lead to early disease interception followed by preventive lifestyle modifications and proactive treatment.
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
| | - 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 Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H D Dainton
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N Jain
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E Belanger
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - A James
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - S George
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - H Weber
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D L Graham
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Schweitzer
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - T B Ladd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Learman
- 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
| | - N P Vanipenta
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - F N Khan
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Mullen
- Advanced Biomedical IT Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - G Shankar
- Advanced Biomedical IT Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S Cook
- Marion County Coroner's Office, Indianapolis, IN, USA
| | - C Humbert
- Marion County Coroner's Office, 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
| | - 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
- 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
|
9
|
Altamura AC, Buoli M, Pozzoli S. Role of immunological factors in the pathophysiology and diagnosis of bipolar disorder: comparison with schizophrenia. Psychiatry Clin Neurosci 2014; 68:21-36. [PMID: 24102953 DOI: 10.1111/pcn.12089] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/05/2013] [Accepted: 05/26/2013] [Indexed: 01/02/2023]
Abstract
Several lines of evidence point to the key role of neurobiological mechanisms and shared genetic background in schizophrenia and bipolar disorder. For both disorders, neurodevelopmental and neurodegenerative processes have been postulated to be relevant for the pathogenesis as well as dysregulation of immuno-inflammatory pathways. Inflammation is a complex biological response to harmful stimuli and it is mediated by cytokines cascades, cellular immune responses, oxidative factors and hormone regulation. Cytokines, in particular, are supposed to play a critical role in infectious and inflammatory processes, mediating the cross-talk between the brain and the immune system; they also possibly contribute to the development of the central nervous system. From this perspective, even though mixed results have been reported, it seems that both schizophrenia and bipolar disorder are associated with an imbalance in inflammatory cytokines; in fact, some of these could represent biological markers of illness and could be possible targets for pharmacological treatments. In light of these considerations, the purpose of the present paper was to provide a comprehensive and critical review of the existing literature about immunological abnormalities in bipolar disorder with particular attention to the similarities and differences with schizophrenia.
Collapse
|
10
|
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: 124] [Impact Index Per Article: 11.3] [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
|
11
|
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: 1.6] [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
|
12
|
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: 305] [Impact Index Per Article: 25.4] [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
|
13
|
Vawter MP, Mamdani F, Macciardi F. An integrative functional genomics approach for discovering biomarkers in schizophrenia. Brief Funct Genomics 2011; 10:387-99. [PMID: 22155586 DOI: 10.1093/bfgp/elr036] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SZ) is a complex disorder resulting from both genetic and environmental causes with a lifetime prevalence world-wide of 1%; however, there are no specific, sensitive and validated biomarkers for SZ. A general unifying hypothesis has been put forward that disease-associated single nucleotide polymorphisms (SNPs) from genome-wide association study (GWAS) are more likely to be associated with gene expression quantitative trait loci (eQTL). We will describe this hypothesis and review primary methodology with refinements for testing this paradigmatic approach in SZ. We will describe biomarker studies of SZ and testing enrichment of SNPs that are associated both with eQTLs and existing GWAS of SZ. SZ-associated SNPs that overlap with eQTLs can be placed into gene-gene expression, protein-protein and protein-DNA interaction networks. Further, those networks can be tested by reducing/silencing the gene expression levels of critical nodes. We present pilot data to support these methods of investigation such as the use of eQTLs to annotate GWASs of SZ, which could be applied to the field of biomarker discovery. Those networks that have association with SNP markers, especially cis-regulated expression, might lead to a more clear understanding of important candidate genes that predispose to disease and alter expression. This method has general application to many complex disorders.
Collapse
Affiliation(s)
- Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry, University of California, Irvine, USA.
| | | | | |
Collapse
|
14
|
Dean B. Dissecting the Syndrome of Schizophrenia: Progress toward Clinically Useful Biomarkers. SCHIZOPHRENIA RESEARCH AND TREATMENT 2011; 2011:614730. [PMID: 22937270 PMCID: PMC3420453 DOI: 10.1155/2011/614730] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 03/28/2011] [Accepted: 04/07/2011] [Indexed: 12/17/2022]
Abstract
The search for clinically useful biomarkers has been one of the holy grails of schizophrenia research. This paper will outline the evolving notion of biomarkers and then outline outcomes from a variety of biomarkers discovery strategies. In particular, the impact of high-throughput screening technologies on biomarker discovery will be highlighted and how new or improved technologies may allow the discovery of either diagnostic biomarkers for schizophrenia or biomarkers that will be useful in determining appropriate treatments for people with the disorder. History tells those involved in biomarker research that the discovery and validation of useful biomarkers is a long process and current progress must always be viewed in that light. However, the approval of the first biomarker screen with some value in predicting responsiveness to antipsychotic drugs suggests that biomarkers can be identified and that these biomarkers that will be useful in diagnosing and treating people with schizophrenia.
Collapse
Affiliation(s)
- Brian Dean
- The Rebecca L. Cooper Research Laboratories, The Mental Health Research Institute, Locked bag 11, Parkville, VIC 3052, Australia
- The Department of Psychiatry, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
15
|
Le-Niculescu H, Case NJ, Hulvershorn L, Patel SD, Bowker D, Gupta J, Bell R, Edenberg HJ, Tsuang MT, Kuczenski R, Geyer MA, Rodd ZA, Niculescu AB. Convergent functional genomic studies of ω-3 fatty acids in stress reactivity, bipolar disorder and alcoholism. Transl Psychiatry 2011; 1:e4. [PMID: 22832392 PMCID: PMC3309466 DOI: 10.1038/tp.2011.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 02/24/2011] [Indexed: 12/28/2022] Open
Abstract
Omega-3 fatty acids have been proposed as an adjuvant treatment option in psychiatric disorders. Given their other health benefits and their relative lack of toxicity, teratogenicity and side effects, they may be particularly useful in children and in females of child-bearing age, especially during pregnancy and postpartum. A comprehensive mechanistic understanding of their effects is needed. Here we report translational studies demonstrating the phenotypic normalization and gene expression effects of dietary omega-3 fatty acids, specifically docosahexaenoic acid (DHA), in a stress-reactive knockout mouse model of bipolar disorder and co-morbid alcoholism, using a bioinformatic convergent functional genomics approach integrating animal model and human data to prioritize disease-relevant genes. Additionally, to validate at a behavioral level the novel observed effects on decreasing alcohol consumption, we also tested the effects of DHA in an independent animal model, alcohol-preferring (P) rats, a well-established animal model of alcoholism. Our studies uncover sex differences, brain region-specific effects and blood biomarkers that may underpin the effects of DHA. Of note, DHA modulates some of the same genes targeted by current psychotropic medications, as well as increases myelin-related gene expression. Myelin-related gene expression decrease is a common, if nonspecific, denominator of neuropsychiatric disorders. In conclusion, our work supports the potential utility of omega-3 fatty acids, specifically DHA, for a spectrum of psychiatric disorders such as stress disorders, bipolar disorder, alcoholism and beyond.
Collapse
Affiliation(s)
- H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Case
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - L Hulvershorn
- 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
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D Bowker
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Gupta
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M T Tsuang
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - R Kuczenski
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - M A Geyer
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - Z A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
16
|
Kurian SM, Le-Niculescu H, Patel SD, Bertram D, Davis J, Dike C, Yehyawi N, Lysaker P, Dustin J, Caligiuri M, Lohr J, Lahiri DK, Nurnberger JI, Faraone SV, Geyer MA, Tsuang MT, Schork NJ, Salomon DR, Niculescu AB. Identification of blood biomarkers for psychosis using convergent functional genomics. Mol Psychiatry 2011; 16:37-58. [PMID: 19935739 DOI: 10.1038/mp.2009.117] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There are to date no objective clinical laboratory blood tests for psychotic disease states. We provide proof of principle for a convergent functional genomics (CFG) approach to help identify and prioritize blood biomarkers for two key psychotic symptoms, one sensory (hallucinations) and one cognitive (delusions). We used gene expression profiling in whole blood samples from patients with schizophrenia and related disorders, with phenotypic information collected at the time of blood draw, then cross-matched the data with other human and animal model lines of evidence. Topping our list of candidate blood biomarkers for hallucinations, we have four genes decreased in expression in high hallucinations states (Fn1, Rhobtb3, Aldh1l1, Mpp3), and three genes increased in high hallucinations states (Arhgef9, Phlda1, S100a6). All of these genes have prior evidence of differential expression in schizophrenia patients. At the top of our list of candidate blood biomarkers for delusions, we have 15 genes decreased in expression in high delusions states (such as Drd2, Apoe, Scamp1, Fn1, Idh1, Aldh1l1), and 16 genes increased in high delusions states (such as Nrg1, Egr1, Pvalb, Dctn1, Nmt1, Tob2). Twenty-five of these genes have prior evidence of differential expression in schizophrenia patients. Predictive scores, based on panels of top candidate biomarkers, show good sensitivity and negative predictive value for detecting high psychosis states in the original cohort as well as in three additional cohorts. These results have implications for the development of objective laboratory tests to measure illness severity and response to treatment in devastating disorders such as schizophrenia.
Collapse
Affiliation(s)
- S M Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bloss CS, Schiabor KM, Schork NJ. Human behavioral informatics in genetic studies of neuropsychiatric disease: multivariate profile-based analysis. Brain Res Bull 2010; 83:177-88. [PMID: 20433907 PMCID: PMC2941546 DOI: 10.1016/j.brainresbull.2010.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 04/17/2010] [Accepted: 04/21/2010] [Indexed: 01/23/2023]
Abstract
While genome-wide association (GWA) studies have yielded notable findings with regard to the identification of risk variants in diseases such as obesity and diabetes, similar studies of schizophrenia - and neuropsychiatric diseases in general - have failed to produce strong findings. One, plausible explanation for this relates to phenotypic heterogeneity and what may be inherent imprecision associated with diagnostic categories in neuropsychiatric disorders. In this review we discuss a general approach to addressing the problem of heterogeneity that draws on concepts in behavioral informatics and the use of multivariable behavioral profiles in genetic studies of neuropsychiatric disease. The use of behavioral profiles as phenotypes eliminates the need for categorizing individuals with different 'subtypes' of a disease into one group and provides a way to investigate genetic susceptibility to different neuropsychiatric disorders that share similar clinical characteristics, such as schizophrenia and bipolar disorder. Further, behavioral profiles are a direct, quantitative representation of the emotional, personality, and neurocognitive functioning of the individuals being studied, and as such, the use of these profiles may provide increased statistical power to detect genetic associations and linkages. We describe and discuss four general data analysis approaches that can be used to analyze and integrate multivariate behavioral profile data and high-dimensional genomic data. Ultimately, we propose that behavioral profile-based phenotypes provide a meaningful alternative to the use of single measures, such as diagnostic category, in genetic association studies of neuropsychiatric disease.
Collapse
Affiliation(s)
- Cinnamon S. Bloss
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
| | - Kelly M. Schiabor
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
| | - Nicholas J. Schork
- Scripps Genomic Medicine, Scripps Translational Science Institute, Scripps Health
- Department of Molecular and Experimental Medicine, The Scripps Research Institute
| |
Collapse
|
18
|
Patel SD, Le-Niculescu H, Koller DL, Green SD, Lahiri DK, McMahon FJ, Nurnberger JI, Niculescu AB. Coming to grips with complex disorders: genetic risk prediction in bipolar disorder using panels of genes identified through convergent functional genomics. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:850-77. [PMID: 20468069 DOI: 10.1002/ajmg.b.31087] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We previously proposed and provided proof of principle for the use of a complementary approach, convergent functional genomics (CFG), combining gene expression and genetic data, from human and animal model studies, as a way of mining the existing GWAS datasets for signals that are there already, but did not reach significance using a genetics-only approach [Le-Niculescu et al., 2009b]. CFG provides a fit-to-disease prioritization of genes that leads to generalizability in independent cohorts, and counterbalances the fit-to-cohort prioritization inherent in classic genetic-only approaches, which have been plagued by poor reproducibility across cohorts. We have now extended our previous work to include more datasets of GWAS, and more recent evidence from other lines of work. In essence our analysis is the most comprehensive integration of genetics and functional genomics to date in the field of bipolar disorder. Biological pathway analyses identified top canonical pathways, and epistatic interaction testing inside these pathways has identified genes that merit future follow-up as direct interactors (intra-pathway epistasis, INPEP). Moreover, we have put together a panel of best P-value single nucleotide polymorphisms (SNPs), based on the top candidate genes we identified. We have developed a genetic risk prediction score (GRPS) based on our panel, and demonstrate how in two independent test cohorts the GRPS differentiates between subjects with bipolar disorder and normal controls, in both European-American and African-American populations. Lastly, we describe a prototype of how such testing could be used to categorize disease risk in individuals and aid personalized medicine approaches, in psychiatry and beyond.
Collapse
Affiliation(s)
- S D Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Niculescu AB, Schork NJ, Salomon DR. Mindscape: a convergent perspective on life, mind, consciousness and happiness. J Affect Disord 2010; 123:1-8. [PMID: 19595463 DOI: 10.1016/j.jad.2009.06.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/03/2009] [Accepted: 06/15/2009] [Indexed: 01/17/2023]
Abstract
What are mind, consciousness and happiness, in the fundamental context of life? We propose a convergent perspective (coupling evolutionary biology, genomics, neurobiology and clinical medicine) that could help us better understand what life, mind, consciousness and happiness are, as well as provides empirically testable practical implications.
Collapse
Affiliation(s)
- Alexander B Niculescu
- Indiana University School of Medicine, Institute of Psychiatric Research, 791 Union Drive, Indianapolis, IN 46202-4887, USA.
| | | | | |
Collapse
|
20
|
Animal models of virus-induced neurobehavioral sequelae: recent advances, methodological issues, and future prospects. Interdiscip Perspect Infect Dis 2010; 2010:380456. [PMID: 20490350 PMCID: PMC2872755 DOI: 10.1155/2010/380456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2009] [Revised: 11/14/2009] [Accepted: 03/09/2010] [Indexed: 01/18/2023] Open
Abstract
Converging lines of clinical and epidemiological evidence suggest that viral infections in early developmental stages may be a causal factor in neuropsychiatric disorders such as schizophrenia, bipolar disorder, and autism-spectrum disorders. This etiological link, however, remains controversial in view of the lack of consistent and reproducible associations between viruses and mental illness. Animal models of virus-induced neurobehavioral disturbances afford powerful tools to test etiological hypotheses and explore pathophysiological mechanisms. Prenatal or neonatal inoculations of neurotropic agents (such as herpes-, influenza-, and retroviruses) in rodents result in a broad spectrum of long-term alterations reminiscent of psychiatric abnormalities. Nevertheless, the complexity of these sequelae often poses methodological and interpretational challenges and thwarts their characterization. The recent conceptual advancements in psychiatric nosology and behavioral science may help determine new heuristic criteria to enhance the translational value of these models. A particularly critical issue is the identification of intermediate phenotypes, defined as quantifiable factors representing single neurochemical, neuropsychological, or neuroanatomical aspects of a diagnostic category. In this paper, we examine how the employment of these novel concepts may lead to new methodological refinements in the study of virus-induced neurobehavioral sequelae through animal models.
Collapse
|
21
|
|
22
|
Niculescu AB, Le-Niculescu H. Convergent Functional Genomics: what we have learned and can learn about genes, pathways, and mechanisms. Neuropsychopharmacology 2010; 35:355-6. [PMID: 20010721 PMCID: PMC3055434 DOI: 10.1038/npp.2009.107] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alexander B Niculescu
- Laboratory of Neurophenomics, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Helen Le-Niculescu
- Laboratory of Neurophenomics, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
23
|
McGrath CL, Glatt SJ, Sklar P, Le-Niculescu H, Kuczenski R, Doyle AE, Biederman J, Mick E, Faraone SV, Niculescu AB, Tsuang MT. Evidence for genetic association of RORB with bipolar disorder. BMC Psychiatry 2009; 9:70. [PMID: 19909500 PMCID: PMC2780413 DOI: 10.1186/1471-244x-9-70] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 11/12/2009] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Bipolar disorder, particularly in children, is characterized by rapid cycling and switching, making circadian clock genes plausible molecular underpinnings for bipolar disorder. We previously reported work establishing mice lacking the clock gene D-box binding protein (DBP) as a stress-reactive genetic animal model of bipolar disorder. Microarray studies revealed that expression of two closely related clock genes, RAR-related orphan receptors alpha (RORA) and beta (RORB), was altered in these mice. These retinoid-related receptors are involved in a number of pathways including neurogenesis, stress response, and modulation of circadian rhythms. Here we report association studies between bipolar disorder and single-nucleotide polymorphisms (SNPs) in RORA and RORB. METHODS We genotyped 355 RORA and RORB SNPs in a pediatric cohort consisting of a family-based sample of 153 trios and an independent, non-overlapping case-control sample of 152 cases and 140 controls. Bipolar disorder in children and adolescents is characterized by increased stress reactivity and frequent episodes of shorter duration; thus our cohort provides a potentially enriched sample for identifying genes involved in cycling and switching. RESULTS We report that four intronic RORB SNPs showed positive associations with the pediatric bipolar phenotype that survived Bonferroni correction for multiple comparisons in the case-control sample. Three RORB haplotype blocks implicating an additional 11 SNPs were also associated with the disease in the case-control sample. However, these significant associations were not replicated in the sample of trios. There was no evidence for association between pediatric bipolar disorder and any RORA SNPs or haplotype blocks after multiple-test correction. In addition, we found no strong evidence for association between the age-at-onset of bipolar disorder with any RORA or RORB SNPs. CONCLUSION Our findings suggest that clock genes in general and RORB in particular may be important candidates for further investigation in the search for the molecular basis of bipolar disorder.
Collapse
Affiliation(s)
- Casey L McGrath
- Department of Psychiatry, Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Stephen J Glatt
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Pamela Sklar
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Helen Le-Niculescu
- Laboratory of Neurophenomics, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Alysa E Doyle
- Pediatric Psychopharmacology Unit, Massachusetts General Hospital; Psychiatric Psychopharmacology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph Biederman
- Pediatric Psychopharmacology Unit, Massachusetts General Hospital; Psychiatric Psychopharmacology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric Mick
- Pediatric Psychopharmacology Unit, Massachusetts General Hospital; Psychiatric Psychopharmacology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen V Faraone
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Alexander B Niculescu
- Laboratory of Neurophenomics, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ming T Tsuang
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Le-Niculescu H, Patel SD, Bhat M, Kuczenski R, Faraone SV, Tsuang MT, McMahon FJ, Schork NJ, Nurnberger JI, Niculescu AB. Convergent functional genomics of genome-wide association data for bipolar disorder: comprehensive identification of candidate genes, pathways and mechanisms. Am J Med Genet B Neuropsychiatr Genet 2009; 150B:155-81. [PMID: 19025758 DOI: 10.1002/ajmg.b.30887] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Given the mounting convergent evidence implicating many more genes in complex disorders such as bipolar disorder than the small number identified unambiguously by the first-generation Genome-Wide Association studies (GWAS) to date, there is a strong need for improvements in methodology. One strategy is to include in the next generation GWAS larger numbers of subjects, and/or to pool independent studies into meta-analyses. We propose and provide proof of principle for the use of a complementary approach, convergent functional genomics (CFG), as a way of mining the existing GWAS datasets for signals that are there already, but did not reach significance using a genetics-only approach. With the CFG approach, the integration of genetics with genomics, of human and animal model data, and of multiple independent lines of evidence converging on the same genes offers a way of extracting signal from noise and prioritizing candidates. In essence our analysis is the most comprehensive integration of genetics and functional genomics to date in the field of bipolar disorder, yielding a series of novel (such as Klf12, Aldh1a1, A2bp1, Ak3l1, Rorb, Rora) and previously known (such as Bdnf, Arntl, Gsk3b, Disc1, Nrg1, Htr2a) candidate genes, blood biomarkers, as well as a comprehensive identification of pathways and mechanisms. These become prime targets for hypothesis driven follow-up studies, new drug development and personalized medicine approaches.
Collapse
Affiliation(s)
- H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Le-Niculescu H, Kurian SM, Yehyawi N, Dike C, Patel SD, Edenberg HJ, Tsuang MT, Salomon DR, Nurnberger JI, Niculescu AB. Identifying blood biomarkers for mood disorders using convergent functional genomics. Mol Psychiatry 2009; 14:156-74. [PMID: 18301394 DOI: 10.1038/mp.2008.11] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There are to date no objective clinical laboratory blood tests for mood disorders. The current reliance on patient self-report of symptom severity and on the clinicians' impression is a rate-limiting step in effective treatment and new drug development. We propose, and provide proof of principle for, an approach to help identify blood biomarkers for mood state. We measured whole-genome gene expression differences in blood samples from subjects with bipolar disorder that had low mood vs those that had high mood at the time of the blood draw, and separately, changes in gene expression in brain and blood of a mouse pharmacogenomic model. We then integrated our human blood gene expression data with animal model gene expression data, human genetic linkage/association data and human postmortem brain data, an approach called convergent functional genomics, as a Bayesian strategy for cross-validating and prioritizing findings. Topping our list of candidate blood biomarker genes we have five genes involved in myelination (Mbp, Edg2, Mag, Pmp22 and Ugt8), and six genes involved in growth factor signaling (Fgfr1, Fzd3, Erbb3, Igfbp4, Igfbp6 and Ptprm). All of these genes have prior evidence of differential expression in human postmortem brains from mood disorder subjects. A predictive score developed based on a panel of 10 top candidate biomarkers (five for high mood and five for low mood) shows sensitivity and specificity for high mood and low mood states, in two independent cohorts. Our studies suggest that blood biomarkers may offer an unexpectedly informative window into brain functioning and disease state.
Collapse
Affiliation(s)
- H Le-Niculescu
- Laboratory of Neurophenomics, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202-4887, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Papiol S, Molina V, Desco M, Rosa A, Reig S, Sanz J, Palomo T, Fañanás L. Gray matter deficits in bipolar disorder are associated with genetic variability at interleukin-1 beta gene (2q13). GENES BRAIN AND BEHAVIOR 2008; 7:796-801. [DOI: 10.1111/j.1601-183x.2008.00421.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Le-Niculescu H, McFarland MJ, Ogden CA, Balaraman Y, Patel S, Tan J, Rodd ZA, Paulus M, Geyer MA, Edenberg HJ, Glatt SJ, Faraone SV, Nurnberger JI, Kuczenski R, Tsuang MT, Niculescu AB. Phenomic, convergent functional genomic, and biomarker studies in a stress-reactive genetic animal model of bipolar disorder and co-morbid alcoholism. Am J Med Genet B Neuropsychiatr Genet 2008; 147B:134-66. [PMID: 18247375 DOI: 10.1002/ajmg.b.30707] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We had previously identified the clock gene D-box binding protein (Dbp) as a potential candidate gene for bipolar disorder and for alcoholism, using a Convergent Functional Genomics (CFG) approach. Here we report that mice with a homozygous deletion of DBP have lower locomotor activity, blunted responses to stimulants, and gain less weight over time. In response to a chronic stress paradigm, these mice exhibit a diametric switch in these phenotypes. DBP knockout mice are also activated by sleep deprivation, similar to bipolar patients, and that activation is prevented by treatment with the mood stabilizer drug valproate. Moreover, these mice show increased alcohol intake following exposure to stress. Microarray studies of brain and blood reveal a pattern of gene expression changes that may explain the observed phenotypes. CFG analysis of the gene expression changes identified a series of novel candidate genes and blood biomarkers for bipolar disorder, alcoholism, and stress reactivity.
Collapse
Affiliation(s)
- H Le-Niculescu
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Le-Niculescu H, McFarland MJ, Mamidipalli S, Ogden CA, Kuczenski R, Kurian SM, Salomon DR, Tsuang MT, Nurnberger JI, Niculescu AB. Convergent Functional Genomics of bipolar disorder: from animal model pharmacogenomics to human genetics and biomarkers. Neurosci Biobehav Rev 2007; 31:897-903. [PMID: 17614132 PMCID: PMC3313450 DOI: 10.1016/j.neubiorev.2007.05.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 05/10/2007] [Accepted: 05/19/2007] [Indexed: 01/12/2023]
Abstract
Progress in understanding the genetic and neurobiological basis of bipolar disorder(s) has come from both human studies and animal model studies. Until recently, the lack of concerted integration between the two approaches has been hindering the pace of discovery, or more exactly, constituted a missed opportunity to accelerate our understanding of this complex and heterogeneous group of disorders. Our group has helped overcome this "lost in translation" barrier by developing an approach called convergent functional genomics (CFG). The approach integrates animal model gene expression data with human genetic linkage/association data, as well as human tissue (postmortem brain, blood) data. This Bayesian strategy for cross-validating findings extracts meaning from large datasets, and prioritizes candidate genes, pathways and mechanisms for subsequent targeted, hypothesis-driven research. The CFG approach may also be particularly useful for identification of blood biomarkers of the illness.
Collapse
Affiliation(s)
- H. Le-Niculescu
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, IN
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - M. J. McFarland
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, IN
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - S. Mamidipalli
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, IN
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - C. A. Ogden
- Drexel University College of Medicine, Philadelphia, PA
| | - R. Kuczenski
- Department of Psychiatry, UC San Diego, La Jolla, CA
| | - S. M. Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - D. R. Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | | | - J. I. Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| | - A. B. Niculescu
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, IN
- INBRAIN, Indiana University School of Medicine, Indianapolis, IN
- Institute of Psychiatric Research, Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
- R. L. Roudebush VA Medical Center, Indianapolis, IN
- Corresponding author, E-mail:
| |
Collapse
|
29
|
Le-Niculescu H, Balaraman Y, Patel S, Tan J, Sidhu K, Jerome RE, Edenberg HJ, Kuczenski R, Geyer MA, Nurnberger JI, Faraone SV, Tsuang MT, Niculescu AB. Towards understanding the schizophrenia code: an expanded convergent functional genomics approach. Am J Med Genet B Neuropsychiatr Genet 2007; 144B:129-58. [PMID: 17266109 DOI: 10.1002/ajmg.b.30481] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Identifying genes for schizophrenia through classical genetic approaches has proven arduous. Here, we present a comprehensive convergent analysis that translationally integrates brain gene expression data from a relevant pharmacogenomic mouse model (involving treatments with a psychomimetic agent - phencyclidine (PCP), and an anti-psychotic - clozapine), with human genetic linkage data and human postmortem brain data, as a Bayesian strategy of cross validating findings. Topping the list of candidate genes, we have three genes involved in GABA neurotransmission (GABRA1, GABBR1, and GAD2), one gene involved in glutamate neurotransmission (GRIA2), one gene involved in neuropeptide signaling (TAC1), two genes involved in synaptic function (SYN2 and KCNJ4), six genes involved in myelin/glial function (CNP, MAL, MBP, PLP1, MOBP and GFAP), and one gene involved in lipid metabolism (LPL). These data suggest that schizophrenia is primarily a disorder of brain functional and structural connectivity, with GABA neurotransmission playing a prominent role. These findings may explain the EEG gamma band abnormalities detected in schizophrenia. The analysis also revealed other high probability candidates genes (neurotransmitter signaling, other structural proteins, ion channels, signal transduction, regulatory enzymes, neuronal migration/neurite outgrowth, clock genes, transcription factors, RNA regulatory genes), pathways and mechanisms of likely importance in pathophysiology. Some of the pathways identified suggest possible avenues for augmentation pharmacotherapy of schizophrenia with other existing agents, such as benzodiazepines, anticonvulsants and lipid modulating agents. Other pathways are new potential targets for drug development. Lastly, a comparison with our earlier work on bipolar disorder illuminates the significant molecular overlap between schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- H Le-Niculescu
- Laboratory of Neurophenomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Schork NJ, Greenwood TA, Braff DL. Statistical genetics concepts and approaches in schizophrenia and related neuropsychiatric research. Schizophr Bull 2007; 33:95-104. [PMID: 17035359 PMCID: PMC2632283 DOI: 10.1093/schbul/sbl045] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Statistical genetics is a research field that focuses on mathematical models and statistical inference methodologies that relate genetic variations (ie, naturally occurring human DNA sequence variations or "polymorphisms") to particular traits or diseases (phenotypes) usually from data collected on large samples of families or individuals. The ultimate goal of such analysis is the identification of genes and genetic variations that influence disease susceptibility. Although of extreme interest and importance, the fact that many genes and environmental factors contribute to neuropsychiatric diseases of public health importance (eg, schizophrenia, bipolar disorder, and depression) complicates relevant studies and suggests that very sophisticated mathematical and statistical modeling may be required. In addition, large-scale contemporary human DNA sequencing and related projects, such as the Human Genome Project and the International HapMap Project, as well as the development of high-throughput DNA sequencing and genotyping technologies have provided statistical geneticists with a great deal of very relevant and appropriate information and resources. Unfortunately, the use of these resources and their interpretation are not straightforward when applied to complex, multifactorial diseases such as schizophrenia. In this brief and largely nonmathematical review of the field of statistical genetics, we describe many of the main concepts, definitions, and issues that motivate contemporary research. We also provide a discussion of the most pressing contemporary problems that demand further research if progress is to be made in the identification of genes and genetic variations that predispose to complex neuropsychiatric diseases.
Collapse
Affiliation(s)
- Nicholas J Schork
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0603, USA.
| | | | | |
Collapse
|
31
|
Niculescu AB. Polypharmacy in oligopopulations: what psychiatric genetics can teach biological psychiatry. Psychiatr Genet 2006; 16:241-4. [PMID: 17106426 DOI: 10.1097/01.ypg.0000242195.74268.f9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Psychiatric genetics and genomics have made major strides in recent years. Some of that knowledge has yet to permeate in the clinical practice of biological psychiatry. The example of cancer-genetics, biology and clinical treatments may be profitable in terms of accelerating translational integration in psychiatry. We propose that current developments in genetics and genomics point to an Early Low-Dose Rational Polypharmacy in Oligopopulations model for psychiatric pharmacotherapy.
Collapse
Affiliation(s)
- Alexander B Niculescu
- Laboratory of Neurophenomics, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
32
|
Rodd ZA, Bertsch BA, Strother WN, Le-Niculescu H, Balaraman Y, Hayden E, Jerome RE, Lumeng L, Nurnberger JI, Edenberg HJ, McBride WJ, Niculescu AB. Candidate genes, pathways and mechanisms for alcoholism: an expanded convergent functional genomics approach. THE PHARMACOGENOMICS JOURNAL 2006; 7:222-56. [PMID: 17033615 DOI: 10.1038/sj.tpj.6500420] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We describe a comprehensive translational approach for identifying candidate genes for alcoholism. The approach relies on the cross-matching of animal model brain gene expression data with human genetic linkage data, as well as human tissue data and biological roles data, an approach termed convergent functional genomics. An analysis of three animal model paradigms, based on inbred alcohol-preferring (iP) and alcohol-non-preferring (iNP) rats, and their response to treatments with alcohol, was used. A comprehensive analysis of microarray gene expression data from five key brain regions (frontal cortex, amygdala, caudate-putamen, nucleus accumbens and hippocampus) was carried out. The Bayesian-like integration of multiple independent lines of evidence, each by itself lacking sufficient discriminatory power, led to the identification of high probability candidate genes, pathways and mechanisms for alcoholism. These data reveal that alcohol has pleiotropic effects on multiple systems, which may explain the diverse neuropsychiatric and medical pathology in alcoholism. Some of the pathways identified suggest avenues for pharmacotherapy of alcoholism with existing agents, such as angiotensin-converting enzyme (ACE) inhibitors. Experiments we carried out in alcohol-preferring rats with an ACE inhibitor show a marked modulation of alcohol intake. Other pathways are new potential targets for drug development. The emergent overall picture is that physical and physiological robustness may permit alcohol-preferring individuals to withstand the aversive effects of alcohol. In conjunction with a higher reactivity to its rewarding effects, they may able to ingest enough of this nonspecific drug for a strong hedonic and addictive effect to occur.
Collapse
Affiliation(s)
- Z A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|