Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Aaron R. Wolen is active.

Publication


Featured researches published by Aaron R. Wolen.


American Journal of Transplantation | 2011

MicroRNA profiles in allograft tissues and paired urines associate with chronic allograft dysfunction with IF/TA

M. J. Scian; Daniel G. Maluf; Krystle G. David; Kellie J. Archer; Jihee L. Suh; Aaron R. Wolen; H. D. Massey; Anne L. King; Todd W.B. Gehr; Adrian H. Cotterell; Mitchell C. Posner; Valeria R. Mas

Despite the advances in immunosuppression, renal allograft attrition over time remains unabated due to chronic allograft dysfunction (CAD) with interstitial fibrosis (IF) and tubular atrophy (TA). We aimed to evaluate microRNA (miRNA) signatures in CAD with IF/TA and appraise correlation with paired urine samples and potential utility in prospective evaluation of graft function. MiRNA signatures were established between CAD with IF/TA versus normal allografts by microarray. Validation of the microarray results and prospective evaluation of urine samples was performed using real‐time quantitative‐PCR (RT‐qPCR). Fifty‐six miRNAs were identified in samples with CAD‐IF/TA. Five miRNAs were selected for further validation based on array fold change, p‐value and in silico predicted mRNA targets. We confirmed the differential expression of these five miRNAs by RT‐qPCR using an independent set of samples. Differential expression was detected for miR‐142‐3p, miR‐204, miR‐107 and miR‐211 (p < 0.001) and miR‐32 (p < 0.05). Furthermore, differential expression of miR‐142‐3p (p < 0.01), miR‐204 (p < 0.01) and miR‐211 (p < 0.05) was also observed between patient groups in urine samples. A characteristic miRNA signature for IF/TA that correlates with paired urine samples was identified. These results support the potential use of miRNAs as noninvasive markers of IF/TA and for monitoring graft function.


PLOS ONE | 2012

Genetic Dissection of Acute Ethanol Responsive Gene Networks in Prefrontal Cortex: Functional and Mechanistic Implications

Aaron R. Wolen; Charles A. Phillips; Michael A. Langston; Alex H. Putman; Paul J. Vorster; Nathan A. Bruce; Timothy P. York; Robert W. Williams; Michael F. Miles

Background Individual differences in initial sensitivity to ethanol are strongly related to the heritable risk of alcoholism in humans. To elucidate key molecular networks that modulate ethanol sensitivity we performed the first systems genetics analysis of ethanol-responsive gene expression in brain regions of the mesocorticolimbic reward circuit (prefrontal cortex, nucleus accumbens, and ventral midbrain) across a highly diverse family of 27 isogenic mouse strains (BXD panel) before and after treatment with ethanol. Results Acute ethanol altered the expression of ∼2,750 genes in one or more regions and 400 transcripts were jointly modulated in all three. Ethanol-responsive gene networks were extracted with a powerful graph theoretical method that efficiently summarized ethanols effects. These networks correlated with acute behavioral responses to ethanol and other drugs of abuse. As predicted, networks were heavily populated by genes controlling synaptic transmission and neuroplasticity. Several of the most densely interconnected network hubs, including Kcnma1 and Gsk3β, are known to influence behavioral or physiological responses to ethanol, validating our overall approach. Other major hub genes like Grm3, Pten and Nrg3 represent novel targets of ethanol effects. Networks were under strong genetic control by variants that we mapped to a small number of chromosomal loci. Using a novel combination of genetic, bioinformatic and network-based approaches, we identified high priority cis-regulatory candidate genes, including Scn1b, Gria1, Sncb and Nell2. Conclusions The ethanol-responsive gene networks identified here represent a previously uncharacterized intermediate phenotype between DNA variation and ethanol sensitivity in mice. Networks involved in synaptic transmission were strongly regulated by ethanol and could contribute to behavioral plasticity seen with chronic ethanol. Our novel finding that hub genes and a small number of loci exert major influence over the ethanol response of gene networks could have important implications for future studies regarding the mechanisms and treatment of alcohol use disorders.


Molecular Psychiatry | 2016

Meta-analysis of genome-wide association studies of anxiety disorders

Takeshi Otowa; Karin Hek; Misun Lee; Enda M. Byrne; Saira Saeed Mirza; Michel G. Nivard; Timothy B. Bigdeli; Steven H. Aggen; Daniel E. Adkins; Aaron R. Wolen; Ayman H. Fanous; Matthew C. Keller; Enrique Castelao; Zoltán Kutalik; S. V. der Auwera; Georg Homuth; Matthias Nauck; Alexander Teumer; Y. Milaneschi; J.J. Hottenga; Nese Direk; A. Hofman; A.G. Uitterlinden; Cornelis L. Mulder; Anjali K. Henders; Sarah E. Medland; S. D. Gordon; A. C. Heath; P. A. F. Madden; M. L. Pergadia

Anxiety disorders (ADs), namely generalized AD, panic disorder and phobias, are common, etiologically complex conditions with a partially genetic basis. Despite differing on diagnostic definitions based on clinical presentation, ADs likely represent various expressions of an underlying common diathesis of abnormal regulation of basic threat–response systems. We conducted genome-wide association analyses in nine samples of European ancestry from seven large, independent studies. To identify genetic variants contributing to genetic susceptibility shared across interview-generated DSM-based ADs, we applied two phenotypic approaches: (1) comparisons between categorical AD cases and supernormal controls, and (2) quantitative phenotypic factor scores (FS) derived from a multivariate analysis combining information across the clinical phenotypes. We used logistic and linear regression, respectively, to analyze the association between these phenotypes and genome-wide single nucleotide polymorphisms. Meta-analysis for each phenotype combined results across the nine samples for over 18 000 unrelated individuals. Each meta-analysis identified a different genome-wide significant region, with the following markers showing the strongest association: for case–control contrasts, rs1709393 located in an uncharacterized non-coding RNA locus on chromosomal band 3q12.3 (P=1.65 × 10−8); for FS, rs1067327 within CAMKMT encoding the calmodulin-lysine N-methyltransferase on chromosomal band 2p21 (P=2.86 × 10−9). Independent replication and further exploration of these findings are needed to more fully understand the role of these variants in risk and expression of ADs.


PLOS ONE | 2016

Time-Course Analysis of Brain Regional Expression Network Responses to Chronic Intermittent Ethanol and Withdrawal: Implications for Mechanisms Underlying Excessive Ethanol Consumption.

Maren L. Smith; Marcelo F. Lopez; Kellie J. Archer; Aaron R. Wolen; Howard C. Becker; Michael F. Miles

Long lasting abusive consumption, dependence, and withdrawal are characteristic features of alcohol use disorders (AUD). Mechanistically, persistent changes in gene expression are hypothesized to contribute to brain adaptations leading to ethanol toxicity and AUD. We employed repeated chronic intermittent ethanol (CIE) exposure by vapor chamber as a mouse model to simulate the cycles of ethanol exposure and withdrawal commonly seen with AUD. This model has been shown to induce progressive ethanol consumption in rodents. Brain CIE-responsive expression networks were identified by microarray analysis across five regions of the mesolimbic dopamine system and extended amygdala with tissue harvested from 0-hours to 7-days following CIE. Weighted Gene Correlated Network Analysis (WGCNA) was used to identify gene networks over-represented for CIE-induced temporal expression changes across brain regions. Differential gene expression analysis showed that long-lasting gene regulation occurred 7-days after the final cycle of ethanol exposure only in prefrontal cortex (PFC) and hippocampus. Across all brain regions, however, ethanol-responsive expression changes occurred mainly within the first 8-hours after removal from ethanol. Bioinformatics analysis showed that neuroinflammatory responses were seen across multiple brain regions at early time-points, whereas co-expression modules related to neuroplasticity, chromatin remodeling, and neurodevelopment were seen at later time-points and in specific brain regions (PFC or HPC). In PFC a module containing Bdnf was identified as highly CIE responsive in a biphasic manner, with peak changes at 0 hours and 5 days following CIE, suggesting a possible role in mechanisms underlying long-term molecular and behavioral response to CIE. Bioinformatics analysis of this network and several other modules identified Let-7 family microRNAs as potential regulators of gene expression changes induced by CIE. Our results suggest a complex temporal and regional pattern of widespread gene network responses involving neuroinflammatory and neuroplasticity related genes as contributing to physiological and behavioral responses to chronic ethanol.


The FASEB Journal | 2016

Sphingosine-1-phosphate phosphatase 2 promotes disruption of mucosal integrity, and contributes to ulcerative colitis in mice and humans

Wei-Ching Huang; Jie Liang; Masayuki Nagahashi; Dorit Avni; Akimitsu Yamada; Michael Maceyka; Aaron R. Wolen; Tomasz Kordula; Sheldon Milstien; Kazuaki Takabe; Tamas Oravecz; Sarah Spiegel

The bioactive sphingolipid sphingosine‐1‐phosphate (S1P) and the kinase that produces it have been implicated in inflammatory bowel diseases in mice and humans; however, little is known about the role of the 2 S1P‐specific phosphohydrolase isoforms, SGPP1 and SGPP2, which catalyze dephosphorylation of S1P to sphingosine. To elucidate their functions, we generated specific knockout mice. Deletion of Sgpp2, which is mainly expressed in the gastrointestinal tract, significantly reduced dextran sodium sulfate (DSS)‐induced colitis severity, whereas deletion of ubiquitously expressed Sgpp1 slightly worsened colitis. Moreover, Sgpp1 deletion enhanced expression of multifunctional proinflammatory cytokines, IL‐6, TNF‐α, and IL‐1β, activation of the transcription factor signal transducer and activator of transcription 3, and immune cell infiltration into the colon. Conversely, Sgpp2‐null mice failed to mount a DSS‐induced systemic inflammatory response. Of interest, Sgpp2 deficiency suppressed DSS‐induced intestinal epithelial cell apoptosis and improved mucosal barrier integrity. Furthermore, down‐regulation of Sgpp2 attenuated LPS‐induced paracellular permeability in cultured cells and enhanced expression of the adherens junction protein E‐cadherin. Finally, in patients with ulcerative colitis, SGPP2 expression was elevated in colitis tissues relative to that in uninvolved tissues. These results indicate that induction of SGPP2 expression contributes to the pathogenesis of colitis by promoting disruption of the mucosal barrier function. SGPP2 may represent a novel therapeutic target in inflammatory bowel disease.—Huang, W.‐C., Liang, J., Nagahashi, M., Avni, D., Yamada, A., Maceyka, M., Wolen, A. R., Kordula, T., Milstien, S., Takabe, K., Oravecz, T., Spiegel, S. Sphingosine‐1‐phosphate phosphatase 2 promotes disruption of mucosal integrity, and contributes to ulcerative colitis in mice and humans. FASEB J. 30, 2945‐2958 (2016). www.fasebj.org


Alcoholism: Clinical and Experimental Research | 2013

Role of Adrenal Glucocorticoid Signaling in Prefrontal Cortex Gene Expression and Acute Behavioral Responses to Ethanol

Blair N. Costin; Aaron R. Wolen; Sylvia Fitting; Keith L. Shelton; Michael F. Miles

BACKGROUND Glucocorticoid hormones modulate acute and chronic behavioral and molecular responses to drugs of abuse including psychostimulants and opioids. There is growing evidence that glucocorticoids might also modulate behavioral responses to ethanol ( EtOH ). Acute EtOH activates the hypothalamic-pituitary-adrenal axis, causing the release of adrenal glucocorticoid hormones. Our prior genomic studies suggest that glucocorticoids play a role in regulating gene expression in the prefrontal cortex (PFC) of DBA2/J (D2) mice following acute EtOH administration. However, few studies have analyzed the role of glucocorticoid signaling in behavioral responses to acute EtOH . Such work could be significant, given the predictive value for the level of response to acute EtOH in the risk for alcoholism. METHODS We studied whether the glucocorticoid receptor (GR) antagonist, RU-486, or adrenalectomy (ADX) altered male D2 mouse behavioral responses to acute (locomotor activation, anxiolysis, or loss-of-righting reflex [LORR]) or repeated (sensitization) EtOH treatment. Whole-genome microarray analysis and bioinformatics approaches were used to identify PFC candidate genes possibly responsible for altered behavioral responses to EtOH following ADX. RESULTS ADX and RU-486 both impaired acute EtOH (2 g/kg)-induced locomotor activation in D2 mice without affecting basal locomotor activity. However, neither ADX nor RU-486 altered the initiation of EtOH sensitization (locomotor activation or jump counts), EtOH -induced anxiolysis, or LORR. ADX mice showed microarray gene expression changes in PFC that significantly overlapped with acute EtOH -responsive gene sets derived by our prior microarray studies. Q-rtPCR analysis verified that ADX decreased PFC expression of Fkbp5 while significantly increasing Gpr6 expression. In addition, high-dose RU-486 pretreatment blunted EtOH -induced Fkbp5 expression. CONCLUSIONS Our studies suggest that EtOH s activation of adrenal glucocorticoid release and subsequent GR activation may partially modulate EtOH s acute locomotor activation in male D2 mice. Furthermore, because adrenal glucocorticoid basal tone regulated PFC gene expression, including a significant set of acute EtOH -responsive genes, this suggests that glucocorticoid-regulated PFC gene expression may be an important factor modulating acute behavioral responses to EtOH .


International Review of Neurobiology | 2010

USING EXPRESSION GENETICS TO STUDY THE NEUROBIOLOGY OF ETHANOL AND ALCOHOLISM

Sean P. Farris; Aaron R. Wolen; Michael F. Miles

Recent simultaneous progress in human and animal model genetics and the advent of microarray whole genome expression profiling have produced prodigious data sets on genetic loci, potential candidate genes, and differential gene expression related to alcoholism and ethanol behaviors. Validated target genes or gene networks functioning in alcoholism are still of meager proportions. Genetical genomics, which combines genetic analysis of both traditional phenotypes and whole genome expression data, offers a potential methodology for characterizing brain gene networks functioning in alcoholism. This chapter will describe concepts, approaches, and recent findings in the field of genetical genomics as it applies to alcohol research.


Molecular Psychiatry | 2016

Meta-analysis of genome-wide association studies of anxiety disorders (vol 21, pg 1391, 2016)

Takeshi Otowa; Karin Hek; Misun Lee; Em Byrne; Saira Saeed Mirza; Michel G. Nivard; Timothy B. Bigdeli; Steven H. Aggen; Daniel E. Adkins; Aaron R. Wolen; Ayman H. Fanous; Matthew C. Keller; Enrique Castelao; Zoltán Kutalik; S. V. der Auwera; Georg Homuth; Matthias Nauck; Alexander Teumer; Y. Milaneschi; J.J. Hottenga; Nese Direk; A. Hofman; A.G. Uitterlinden; C L Mulder; Ak Henders; S. E. Medland; S. D. Gordon; A. C. Heath; P. A. F. Madden; M. L. Pergadia

Correction to: Molecular Psychiatry (2015); advance online publication 12 January 2016; doi:10.1038/mp.2015.197 Following publication of the above article, the authors noticed that the fifteenth author’s name was presented incorrectly. The author’s name should have appeared as S Van der Auwera. The publisher regrets the error.


Addiction | 2015

Genomic influences on alcohol problems in a population-based sample of young adults

Alexis C. Edwards; Fazil Aliev; Aaron R. Wolen; Jessica E. Salvatore; Charles O. Gardner; George McMahon; David Evans; John Macleod; Matthew Hickman; Danielle M. Dick; Kenneth S. Kendler

AIMS Alcohol problems (AP) contribute substantially to the global disease burden. Twin and family studies suggest that AP are genetically influenced, although few studies have identified variants or genes that are robustly associated with risk. This study identifies genetic and genomic influences on AP during young adulthood, which is often when drinking habits are established. DESIGN We conducted a genome-wide association study of AP. We further conducted gene-based tests, gene ontology analyses and functional genomic enrichment analyses to assess genomic factors beyond single variants that are relevant to AP. SETTING The Avon Longitudinal Study of Parents and Children, a large population-based study of a UK birth cohort. PARTICIPANTS Genetic and phenotypical data were available for 4304 participants. MEASUREMENTS The AP phenotype was a factor score derived from items from the Alcohol Use Disorders Identification Test, symptoms of DSM-IV alcohol dependence, and three additional problem-related items. FINDINGS One variant met genome-wide significance criteria. Four out of 22,880 genes subjected to gene-based analyses survived a stringent significance threshold (q < 0.05); none of these have been implicated previously in alcohol-related phenotypes. Several biologically plausible gene ontologies were statistically over-represented among implicated single nucleotide polymorphisms (SNPs). SNPs on the Illumina 550 K SNP chip accounted for ~5% of the phenotypical variance in AP. CONCLUSIONS Genetic and genomic factors appear to play a role in alcohol problems in young adults. Genes involved in nervous system-related processes, such as signal transduction and neurogenesis, potentially contribute to liability to alcohol problems, as do genes expressed in non-brain tissues.


Clinical Epigenetics | 2016

Establishing an analytic pipeline for genome-wide DNA methylation.

Michelle L. Wright; Mikail G. Dozmorov; Aaron R. Wolen; Colleen Jackson-Cook; Angela Starkweather; Debra E. Lyon; Timothy P. York

The need for research investigating DNA methylation (DNAm) in clinical studies has increased, leading to the evolution of new analytic methods to improve accuracy and reproducibility of the interpretation of results from these studies. The purpose of this article is to provide clinical researchers with a summary of the major data processing steps routinely applied in clinical studies investigating genome-wide DNAm using the Illumina HumanMethylation 450K BeadChip. In most studies, the primary goal of employing DNAm analysis is to identify differential methylation at CpG sites among phenotypic groups. Experimental design considerations are crucial at the onset to minimize bias from factors related to sample processing and avoid confounding experimental variables with non-biological batch effects. Although there are currently no de facto standard methods for analyzing these data, we review the major steps in processing DNAm data recommended by several research studies. We describe several variations available for clinical researchers to process, analyze, and interpret DNAm data. These insights are applicable to most types of genome-wide DNAm array platforms and will be applicable for the next generation of DNAm array technologies (e.g., the 850K array). Selection of the DNAm analytic pipeline followed by investigators should be guided by the research question and supported by recently published methods.

Collaboration


Dive into the Aaron R. Wolen's collaboration.

Top Co-Authors

Avatar

Michael F. Miles

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Timothy P. York

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Danielle M. Dick

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Fazil Aliev

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jessica E. Salvatore

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Alexis C. Edwards

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Amy L. Olex

Wake Forest University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeanne E. Savage

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Kenneth S. Kendler

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge