Skip to main content
  • Research article
  • Open access
  • Published:

Transcriptome profiling and pathway analysis of genes expressed differentially in participants with or without a positive response to topiramate treatment for methamphetamine addiction

Abstract

Background

Developing efficacious medications to treat methamphetamine dependence is a global challenge in public health. Topiramate (TPM) is undergoing evaluation for this indication. The molecular mechanisms underlying its effects are largely unknown. Examining the effects of TPM on genome-wide gene expression in methamphetamine addicts is a clinically and scientifically important component of understanding its therapeutic profile.

Methods

In this double-blind, placebo-controlled clinical trial, 140 individuals who met the DSM-IV criteria for methamphetamine dependence were randomized to receive either TPM or placebo, of whom 99 consented to participate in our genome-wide expression study. The RNA samples were collected from whole blood for 50 TPM- and 49 placebo-treated participants at three time points: baseline and the ends of weeks 8 and 12. Genome-wide expression profiles and pathways of the two groups were compared for the responders and non-responders at Weeks 8 and 12. To minimize individual variations, expression of all examined genes at Weeks 8 and 12 were normalized to the values at baseline prior to identification of differentially expressed genes and pathways.

Results

At the single-gene level, we identified 1054, 502, 204, and 404 genes at nominal P values < 0.01 in the responders vs. non-responders at Weeks 8 and 12 for the TPM and placebo groups, respectively. Among them, expression of 159, 38, 2, and 21 genes was still significantly different after Bonferroni corrections for multiple testing. Many of these genes, such as GRINA, PRKACA, PRKCI, SNAP23, and TRAK2, which are involved in glutamate receptor and GABA receptor signaling, are direct targets for TPM. In contrast, no TPM drug targets were identified in the 38 significant genes for the Week 8 placebo group. Pathway analyses based on nominally significant genes revealed 27 enriched pathways shared by the Weeks 8 and 12 TPM groups. These pathways are involved in relevant physiological functions such as neuronal function/synaptic plasticity, signal transduction, cardiovascular function, and inflammation/immune function.

Conclusion

Topiramate treatment of methamphetamine addicts significantly modulates the expression of genes involved in multiple biological processes underlying addiction behavior and other physiological functions.

Peer Review reports

Background

Methamphetamine (METH), an N-methyl derivative of amphetamine commonly abused recreationally, is a powerfully addictive psychostimulant that affects the central nervous system (CNS) dramatically [1]. Dependence on the drug has risen to an epidemic level worldwide [2], and in 2007, 529,000 Americans (ca. 0.2% of the US population) were METH users [3].

METH induces long-term changes in behavior, including sensitization and dependence [4],[5], as well as deficits in cognitive function [6]-[8], and causes psychiatric symptoms such as hallucinations and delusions [9]. Its use and abuse have been associated with several significant health risks, including cardiac dysrhythmia, stroke, high blood pressure, hyperthermia, and CNS abnormalities [10],[11] that are thought to reflect changes in the signaling and metabolism of neurotransmitters such as dopamine, serotonin, and glutamate [10],[12]-[16].

Unfortunately, no efficacious medication for METH dependence has been developed to date [17]. There is a great need, not only for novel treatments, but for understanding of its molecular mechanisms. Topiramate (TPM), a sulfamate-substituted derivative of the monosaccharide D-fructose [18], has been efficacious in the treatment of alcohol dependence [19] and in promoting smoking cessation among alcohol-dependent smokers [20]. A preliminary study suggests that it also may be useful for treating cocaine dependence [21]. These therapeutic effects have been attributed to its hypothesized potential to reduce the release of cortico-mesolimbic dopamine, the neurotransmitter primarily responsible for the acquisition and maintenance of drug-seeking behaviors for the majority of abused drugs, including amphetamines. Thus, TPM might be efficacious for treating METH dependence [22]. However, the effects of TPM on METH-dependent subjects seem to be complex. Whereas earlier studies have not uncovered any deleterious interactions between TPM and METH with respect to cognitive performance, attention, or concentration, TPM tends to enhance METH-induced increases in attention and decrease perceptual-motor function [22]. Also, TPM accentuates markedly the positive subjective effects of METH, although not craving or reinforcement [23]. Although several hypotheses have been offered on the basis of clinical laboratory studies for the effects of TPM on METH dependence [22]-[25], the molecular mechanisms remain unclear.

In a recently completed double-blind, multi-center, placebo-controlled clinical trial of the treatment of METH dependence with TPM, mixed results were obtained [26]. Thus, although TPM did not increase abstinence from METH use, it significantly reduced urine METH concentrations and observer-rated severity of dependence [26]. From this trial, a genome-wide expression analysis was conducted on RNA extracted from the blood of participants, with the goal of identifying differentially expressed genes and pathways in the responders and non-responders. Such a global gene expression investigation not only provides evidence at the molecular level explaining the interaction of TPM and METH but also may help us to evaluate the pharmacological effect of TPM on METH dependence.

Results

Grouping study participants used for transcriptome analysis

On the basis of 209 chips that passed quality control, 49 participants in the placebo group and 50 in the TPM group were included. According to the criteria for primary efficacy outcome [26] (also see Methods), these participants were classified as responders or non-responders. For these participants, only 43 had a gene expression study at all three time points, 27 and 24 of which could be classified as responders or non-responders, respectively. For the other 16 participants, either no valid urine samples were tested or the patients were excluded for other reasons at Weeks 8 and 12 (see Additional file 1: Figure S1). To increase the sample size, we included some participants having valid gene expression data at Week 8 but not at Week 0 (baseline) among the Week 8 samples, as well as those participants with valid gene expression data at both Weeks 8 and 12 but not at baseline. Finally, we identified 5 responders and 17 non-responders in the Week 8 TPM group, 4 responders and 17 non-responders in the Week 8 placebo group (see Additional file 1: Figure S1A), 6 responders and 11 non-responders in the Week 12 TPM group, and 2 responders and 13 non-responders in the Week 12 placebo group (see Additional file 1: Figure S1B).

Identification of genes differentially expressed in responders and non-responders at Weeks 8 and 12

At a significance level of 0.01, we identified 1,054 (FDR: 0.009 ± 0.010; range <1 × 10−5 - 0.035), 502 (FDR: 0.027 ± 0.021; range: <1 × 10−5 - 0.070), 204 (FDR: 0.113 ± 0.034; range: 0.003 - 0.160), and 404 (FDR: 0.033 ± 0.024; range: <1 × 10−5 - 0.084) differentially expressed genes between responders and non-responders for the Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo groups, respectively (see Additional file 2: Tables S1-S4 for details). Of these four groups, the Week 8 TPM group had the lowest FDR. To take into account the number of genes tested in the four groups, 159, 38, 2, and 21 genes, respectively, remained significant at Bonferroni-corrected P values < 0.05.

In the Week 8 TPM group, 159 genes were significantly changed with a Bonferroni-corrected P value of < 0.05, with 97 being up-regulated and 62 down-regulated comparing positive and negative responders. Importantly, none of these 159 genes overlapped with the 38 genes detected in the Week 8 placebo group at a Bonferroni-corrected P value of 0.05 (Additional file 2: Table S2). Tables 1 and 2 show, respectively, the representative up-regulated and down-regulated genes whose functions are related to cell adhesion/motion, nervous system development and function/synaptic plasticity, signal transduction, ubiquitination/intracellular protein transport, mitochondrial function/metabolism and energy pathways, and immune system function categories.

Table 1 A list of 48 representative genes significantly up-regulated in week 8 topiramate group a
Table 2 A List of 45 representative genes significantly down-regulated in week 8 topiramate group a

In the Week 12 TPM group, we detected only two genes, ITCH and MKNK2, whose expression remained significant after Bonferroni correction for multiple testing (Additional file 2: Table S3). Both were down-regulated by TPM and are in the nervous system development and function/synaptic plasticity category. Although the exact reason is unknown, we suspect that the small size of the Week 12 TPM group might have contributed. None of them overlapped with those 21 genes changed by placebo at Week 12 with Bonferroni-corrected P values < 0.05 (Additional file 2: Table S4).

Pathways identified by IPA

The differentially expressed genes were subjected to pathway analysis using the IPA. A total of 114, 41, 54, and 25 pathways with at least three genes overexpressed were enriched at a nominal P value of < 0.05 between responders and non-responders for Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo, respectively. Among these pathways, 21 significantly enriched pathways with an FDR of < 0.05 at either time point or FDR < 0.10 at both time points were shared exclusively by the Week 8 and Week 12 TPM groups (Table 3), suggesting they are more likely to be the pathways related to the treatment effect of TPM in METH-dependent subjects. No significantly enriched pathways were shared exclusively by the Week 8 and Week 12 placebo groups, with FDRs < 0.10 at both time points. Although 163, 149, 137, and 120 pathways were detected for the Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo groups, respectively, at a significance level of 0.05, only 46, 5, 6, and 0 pathways remained significant after Bonferroni correction for multiple testing. A comparison of these significant pathways after correction for multiple testing revealed that only two pathways (i.e., B-cell receptor signaling and renin-angiotensin signaling) were shared exclusively by the Week 8 and Week 12 TPM groups, and no pathways were shared by the Week 8 and Week 12 placebo groups.

Table 3 Significantly enriched pathways detected exclusively in week 8 and week 12 topiramate groups (n = 27)a

Pathways identified by onto-tools pathway-express

Next, we performed pathway analysis on the nominally significantly expressed genes using Onto-Tools Pathway-Express. A total of 47, 21, 32, and 25 KEGG pathways with at least three overexpressed genes were enriched at nominal P values < 0.05 between responders and non-responders for the Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo groups, respectively. Among them, eight significantly enriched KEGG pathways with FDRs < 0.05 at either time point or FDRs < 0.10 at both time points were shared exclusively by the Week 8 and Week 12 TPM groups (Table 3). Comparing the pathways detected by Onto-tools with those detected by IPA, we found three were shared: synaptic long-term potentiation, Fc epsilon RI signaling, and natural killer-cell signaling. In contrast, no significantly enriched KEGG pathways were shared by the Week 8 and Week 12 placebo groups, with FDRs < 0.10 at both time points. Again, although 81, 64, 60, and 65 pathways were detected for the Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo groups, after Bonferroni correction, only 19, 5, 3, and 5 pathways remained significant. Furthermore, only two pathways (i.e., MAPK signaling and T-cell receptor signaling) were shared exclusively by the Week 8 and Week 12 TPM groups, and no pathways were shared by the Week 8 and Week 12 placebo groups.

Combining the results of IPA and Onto-Tools Pathway-Express, at the nominal P values < 0.05 and further restricting by FDRs < 0.05 at either time point or < 0.10 at both, a total of 27 pathways were identified (see Table 3). These pathways are involved in a spectrum of physiological functions: some are associated mainly with signal transduction (Fc epsilon RI signaling, LPS-stimulated MAPK signaling, p38 MAPK signaling, and SAPK/JNK signaling), whereas others are related to cardiovascular function (cardiac hypertrophy signaling, and renin-angiotensin signaling), and inflammation/immune function (B-cell activating-factor signaling, CCR3 signaling in eosinophils, CCR5 signaling in macrophages, chemokine signaling, CXCR4 signaling, epithelial cell signaling in Helicobacter pylori infection, natural killer cell signaling, and role of PKR in interferon induction and antiviral response).

The essential pathways related to neuronal function/synaptic plasticity include alpha-adrenergic signaling, ephrin receptor signaling, ErbB signaling, FGF signaling, GnRH signaling, mTOR signaling, neurotrophin/TRK signaling, and synaptic long-term potentiation. The genes in the synaptic long-term potentiation pathway that were changed by TPM at Week 8 and Week 12 are depicted in Figure 1.

Figure 1
figure 1

Enriched synaptic long-term potentiation canonical pathway , identified by ingenuity pathway analysis based on differentially expressed genes ( P value<0.05 ) with the ordinary students t -test. The pathway was also detected by onto-tools pathway-express. (A) Week 8 TPM group (29 genes: ATF2, CAMK2D, CAMK2G, CREB1, EP300, GNAQ, GRINA, MAP2K1, MAPK1, MAPK3, PLCB2, PPP1CA, PPP1CB, PPP1CC, PPP1R10, PPP1R12A, PPP1R14B, PPP1R7, PPP3CB, PPP3CC, PRKACA, PRKACB, PRKAR1A, PRKCD, PRKCH, PRKCI, PRKCQ, PRKCZ, and RRAS); and (B) Week 12 TPM group (10 genes; ATF4, CREB5, EP300, GNAQ, KRAS, PPP1R10, PRKACB, PRKAR2A, PRKCB, and PRKCQ). Symbols with a single border represent single genes; those with a double border represent complexes of genes or the possibility that alternative genes might act in the pathway. Red symbols represent up-regulated gene clusters and green symbols represent down-regulated clusters.

Discussion

The current study is the first genome-wide expression investigation into the effects of TPM for the treatment of METH dependence. By profiling genome-wide expression patterns in human white blood cells from METH-dependent subjects who received either oral TPM or placebo, we identified various number of genes that are differentially expressed between responders and non-responders in the TPM-treated and placebo control groups. Further clustering of these altered genes according to their function revealed the significantly enriched pathways governing neuroplasticity and neurotoxicity/neurodegeneration (see Figure 2). Given the primary purpose of this clinical trial, in this discussion, we focus primarily on how TPM may regulate molecular pathways of synaptic plasticity underlying METH’s reward and reinforcing effects that influence abstinence.

Figure 2
figure 2

Integrated model of the biological pathways related to TPM treatment for methamphetamine addiction. The joint effects of TPM and methamphetamine act on multiple molecular pathways that eventually result in modulations of neuroplasticity and neurotoxicity/neurodegeneration, which have a combined effect on cognitive/behavioral function. Pathways enriched exclusively in the TPM responder groups at Weeks 8 and 12 are highlighted in gray.

Exposure to drugs of abuse triggers various gene expression changes resulting in complex neural adaptations that determine the addictive properties of abusive drugs [27]. Among these changes, modifications in long-term synaptic potentiation (LTP) of neuroplasticity are fundamental in instilling reward and reinforcing the drug effects [28]. With evidence from decades of molecular research, it is established that METH alters LTP through activation of dopamine or glutamate surface receptors or both [29] that are linked to the intracellular signal transduction extracellular-signal-regulated-kinase (ERK) pathway. Among the surface neurotransmitter receptors regulating this pathway, the only receptor gene we found to be differentially expressed in the responders and non-responders to TPM was ionotropic glutamate receptor N-methyl D-aspartate-associated protein 1 (GRINA). This protein is a subtype of N-methyl D-aspartate (NMDA) receptors that are antagonized by TPM [30],[31] and activated by METH [32],[33]. In the TPM responder group, GRINA was down-regulated at Week 8, implying fewer NMDA receptors at the synapse. Although expression of none of the other primary target genes of TPM (such as GABAA and AMPA/kainate glutamate receptors) was altered by TPM, three genes coding for membrane trafficking proteins (DLG1, SNAP23, and TRAK2) associated with these receptors were up-regulated in TPM responders compared with non-responders and placebo-treated subjects. The DLG1 gene encodes a synaptic scaffolding protein (also known as synapse-associated protein 97) involved in synapse formation [34] and trafficking of AMPA [35],[36], kainate [37], and NMDA [38],[39] glutamate receptors. SNAP23 is a scaffolding protein that aids in stabilizing NMDA receptors at the neuronal surface [40]. The TRAK2 product is involved in GABAB-receptor trafficking [41]. In rat neocortex, DLG1 mRNA is up-regulated by the NMDA antagonist phencyclidine but not by METH [42]. Considering these factors, it is possible that TPM-associated alterations in the expression of AMPA, kainite, and GABAA receptors at the neuronal surface are more likely to be further governed by post-transcriptional modifications such as receptor phosphorylation and trafficking to and from the synaptic membrane, rather than through alterations in their transcription.

Once METH activates its neural receptors, they activate ERK via the upstream cytoplasmic regulators of the ERK pathway; activated ERK translocates from the cytoplasm to the nucleus and phosphorylates cAMP response element binding protein (CREB) [27] to facilitate METH-induced gene expression, serving as the mediator between the nucleus and the target receptors of METH at the neuronal surface [43]. The drug might activate the ERK pathway via either up-regulation of gene transcription, post-transcriptional activation of protein phosphorylation, or both. In the present study, expression of several genes of the ERK pathway was down-regulated in responders at Week 8 of TPM treatment compared with non-responders and the placebo-treated subjects (see Figure 1A), suggesting a “reversal” of METH-induced up-regulation of ERK pathway genes. At Week 8, these TPM-related down-regulated genes included ERK-1 (MAPK3) and its upstream regulators, protein kinase A (PRKACA), protein kinases C and Z (PRKCD and PRKCZ), Ras-related genes (ARHGEF2, RHOT2, and RRAS), and EP300, which encodes a transcriptional co-activator that forms a complex with CREB-binding protein (CBP). By Week 12, besides EP300, the transcription factor CREB gene CREB5 expression was down-regulated in TPM responders.

Given these findings, it is reasonable to hypothesize that (1) reversal of ERK and CREB over-expression results in blocking of METH-dependent transcription activity and consequently disruption of METH-induced LTP and (2) non-responders may harbor variants that affect expression of genes that are down-regulated in TPM responders. These possibilities have gained support from several lines of evidence reported by other investigators. For example, Narita et al. [29] demonstrated that blockade of protein kinase C (PKC) abolishes behavioral sensitization to METH. Human laboratory studies have indicated a partial inhibition of METH’s reinforcing effects by TPM at the same dosage used in the current study [23]. More importantly, our findings corroborate the concept that TPM would be a possible treatment for METH addiction through facilitating the inhibitory effects of GABA and blocking glutamate excitatory effects on dopamine neurons [22],[23].

Apart from pathways governing neural plasticity, the functional category with the largest number of affected pathways was in immune function (see Table 3). Data on TPM’s effects on immune mediators is sparse, with a few studies emerging recently. Among the ten immune function-related pathways detected in the current study, only the T-cell receptor signaling pathway has been reported previously to be regulated by TPM [44]. A common feature of all these pathways and the pathways governing neuroplasticity is their use of the mitogen-activated-protein-kinase (MAPK) pathway as the central component. As this is not the primary focus of this report, a detailed discussion of those immune-related pathways will not be provided here.

The reliability of our findings is strengthened by a number of aspects of our study design: First, the present study included both a positive (TPM non-responders) and a negative (placebo) control group. Inclusion of a placebo group provided us with a reference necessary for the exploration of gene expression alterations induced specifically by TPM rather than by the absence or reduction of METH use or any other non-specific factors. For example, EP300, a member of the CREB gene family, was down-regulated in both the Weeks 8 and 12 TPM responder and the Week 12 placebo responder groups, suggesting that the regulatory effect of the gene is not specific to TPM, whereas CREB5, discussed above, was down-regulated only in Week 12 TPM responders, suggesting a TPM-specific effect. Further, the inclusion of a positive control group aided us in identifying genes and pathways associated with METH abstinence, which was the primary outcome of this clinical study. Second, we analyzed expression data from three time points, namely, the baseline (prior to starting TPM treatment) and Weeks 8 and 12 for each individual. This approach allowed us to correct for any confounding effects that might be caused by significant individual gene expression differences among subjects at baseline, by normalizing the extent of expression at Weeks 8 and 12 with the patient’s own baseline expression and increasing the reliability of the findings by utilizing Week 12 expression patterns to confirm those that occurred at Week 8. The third main strength of the present study is that the dose of TPM administered throughout the treatment period was well within the drug’s therapeutic range [22],[23], and therefore, we can confidently conclude that the TPM-dependent expression alterations we detected were not related to TPM’s toxic effects, but rather to its therapeutic effects. Finally, we believe that, with the level of rigorousness of the clinical and statistical criteria employed in defining treatment responders and significantly altered genes and pathways, the chance that our findings are falsely positive is minimal.

However, this study is limited by several factors, of which the most notable is the small sample size for some comparison groups. Especially, the number of responders in the TPM and placebo groups were not balanced for either Week 8 or Week 12 (4 and 2 subjects for Weeks 8 and 12 in the placebo group vs. 5 and 6 subjects in TPM group for Weeks 8 and 12). However, these numbers are not distinctly smaller than those in other pharmacogenomic/expression studies published in the literature [45] and provided us with an 85% statistical power to draw conclusions about individual genes and pathways [46]. On the other hand, it could be argued that the imbalance in the number of responders in the two groups was attributable in part to weaker effects of the placebo in promoting abstinence compared with TPM. Because of the small samples, we did not consider covariate effects such as age, sex, and ethnicity in assessing single-gene effects. Although we believe the results obtained from such samples are reliable, extra attention should be paid in interpreting the expression pattern of single genes, especially those identified from the placebo groups. Another main limitation of our study is that we used a peripheral white blood cell model to study the gene expression alterations associated with neuronal functions. Peripheral blood is an easily accessible source of RNA for analysis of environmental exposure and disease conditions [47]-[49]. Circulating leukocytes can be used to infer gene expression in other tissues [50]. Indeed, constituents of blood maintain the balance of homeostasis, modulate immunity or inflammation, partake in stress signaling, and facilitate cellular communication in vascular-associated tissues, including those of the CNS [51]. Sullivan et al. [52] conducted a secondary data analysis of transcriptional profiling of 79 diverse human tissues and found that whole blood shared substantial gene expression similarities with multiple brain tissues such as the amygdala, caudate nucleus, prefrontal cortex, and whole brain (the median Spearman correlation coefficient for the group was 0.52), indicating that gene expression in whole blood can be a robust and valid surrogate for gene expression in the brain [52]. However, in another recent study, only weak correlation was detected between gene expression in the brain and that in blood samples [53]. Under such conditions, although the gene expression data from whole blood may provide useful information to infer the biological processes underlying the interaction of TPM and METH in the neuronal system, more direct evidence obtained from brain tissues is necessary in order to verify the findings reported in this study.

Conclusions

In summary, with application of rigorous clinical and statistical criteria, we demonstrated that TPM mitigates METH’s reinforcing effects, possibly through reversal of some of the dysregulated genes in pathways governing synaptic plasticity to their normal state. Further studies are necessary to replicate these findings as well as to identify genetic variations that may have resulted in regulatory differences observed in TPM responders vs. non-responders. Identification of such molecular mechanisms will help greatly in developing efficacious medications for the treatment of METH dependence.

Methods

Study design and blood sample collection

This was a double-blind, multi-center, placebo-controlled, randomized, parallel-group study for METH-dependent outpatients [26]. Under the inter-agency agreement between the National Institute on Drug Abuse and the Veterans Affairs (VA) Cooperative Programs, eight medical centers participated. The sites’ Institutional Review Boards and the VA Human Rights Committee approved the protocol for and conduct of the study.

Subjects meeting the eligibility criteria after a 14-day screening period and a baseline assessment were randomized into equivalent-size groups for oral treatment with TPM or placebo daily for 91 days. There was a dose titration phase (Days 1 to 35) to a maximum tolerated dose of TPM not to exceed 200 mg/day, a maintenance phase (Days 36 to 84), and a taper phase (Days 85 to 91). To continue in the study, subjects had to maintain a minimum daily dose of 50 mg. Blood samples were collected on Day 1 (considered the baseline) and at the end of Weeks 8 and 12 from every participant who consented to participate in the genetics/expression study. The rationale for using weeks 8 and 12 of TPM treatment in the genetic/expression study was that these two time points were in the middle of the maintenance phase of the maximum dose for each patient and the end of treatment, respectively. At the two time points, because the TPM dose given to each patient became relatively stable, this would reduce variability of drugs received among patients, thus likely increasing statistical power of identifying differentially expressed genes and pathways. All blood samples for this study were collected in PAXgeneTM blood tubes using standard phlebotomy technique.

Primary efficacy outcome measure

The primary efficacy outcome measure was METH use or non-use during each week of the entire period from weeks 1 to 12. For each participant, urine samples were collected three times per week. A positive use week was defined as any week in which at least one of the urine tests was positive for METH and a negative use week as one in which all three tests were negative. The value was considered to be missing if no urine sample was collected. On the basis of the primary efficacy outcome measures for the entire trial period, each study participant in either the TPM or the placebo group was classified as either a positive or negative responder to the treatment, which was referred to as responder or non-responder in this study. For example, a TPM responder at Week 8 means for this participant receiving TPM treatment no METH was detected in all the three urine samples for Week 8 (negative use week); whereas for a TPM non-responder, METH was detected in one or more urine samples. Our aim was to determine which genes were differentially expressed in the responders and non-responders of the TPM or placebo group during a given week. Because we collected blood samples from each participant at baseline and Weeks 8 and 12, we formed four analysis groups: Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo, according to the positive or negative use information at Weeks 8 and 12, respectively. Because not all participants contributed blood samples at both time points, the final sample sizes were different for each group.

RNA isolation and gene expression analysis

Blood samples were collected at approximately the same time of day for each participant for all three time points to control for potential circadian rhythm effects on gene expression. Total RNA was extracted using the PAXgene™ Blood RNA Isolation Kit (Qiagen, Valencia, CA, USA). Genome-wide expression of each sample was assessed with a Human Genome U-133 Plus 2.0 array (Affymetrix Inc., Santa Clara, CA, USA) by Expression Analysis Inc. (Durham, NC). Briefly, the double-stranded cDNA was used in a T7 RNA polymerase in vitro transcription reaction (Ambion, Austin, TX, USA) containing biotin-labeled ribonucleotides CTP and UTP. The resulting labeled cRNAs were then hybridized to HG-U133plus2.0 arrays.

Quality control and bioinformatics analysis of array data

Outlier array detection and quality assessment: In total, there were 212 HG-U133plus2.0 arrays from 99 study participants, which included 91 arrays at baseline, 65 at Week 8, and 56 at Week 12. The 212 “.CEL” files generated by the Microarray Suite (MAS 5.0; Affymetrix) were converted into “.DCP” files using dChip 2008 software (http://biosun1.harvard.edu/~cli/dchip_2008_05.exe). We used the “% array outlier” diagnostic metric to detect outlier arrays, defined as the percentage of outlier probe sets in one array [54]. If this percentage exceeded 5%, the array was called an “outlier.” Three arrays at baseline were found to have a “% array outlier” metric > 5% and were excluded from further analysis. For quality assessment of the remaining 209 chips, the distributions of log2-transformed raw probe-level intensities were visualized by boxplots, and no anomalies were found (data not shown).

Data pre-processing and normalization: Data quality assessment was followed by data pre-processing and normalization with the Robust Multi-Array Average (RMA) algorithm [55], implemented in the RMA function in the Bioconductor Affy package [56]. The RMA is a statistical method comprising three procedures performing the following functions: (i) convolution background correction; (ii) probe-level quantile normalization; and (iii) median polish summarization for each probe set to estimate the log2 scale expression values. A matrix of expression values was computed for the 209 “.CEL” files. The expression values after normalization were similar across arrays.

Probe set filtering: The HG-U133plus2.0 array contains 54,675 oligonucleotide-based probe sets. However, not all of these sets correspond to well-defined genes. By using the latest Affymetrix annotation file (dated November 30, 2008), we found that a total of 33,752 (61.73%) probe sets correspond to unique genes, whereas the remaining probe sets do not and were thus excluded from our statistical analysis. Furthermore, we implemented a series of filtering procedures to reduce the number of probe sets to be tested, which is summarized as follows: (i) FilteringAbsence callprobe sets: We applied a Bioconductor package called “Presence-Absence Calls with Negative Probesets” (PANP) that uses Affymetrix-reported probe sets with no known hybridization partners. PANP uses a simple empirically derived approach to generate P values for thresholds to define “presence/absence” calls. The “presence/absence” calls and P values are returned as two matrices: “Pcalls” and “Pvals,” respectively. Probe sets with < 50% present calls among all arrays within each group were removed, which is considered restrictive [57],[58], leaving ~15,000 probe sets for further analysis. (ii) Filtering biologically irrelevant genes and duplicate probe set(s) for each selected gene: Among the ~15,000 probe sets, control sets of various housekeeping genes (e.g., GAPDH) and spiked-in controls (e.g., Ec-bioB, Ec-bioC, Ec-bioD), as well as those genes that are not well defined or have unknown functions were removed. After removing duplicate probe set(s) for the same gene, such that only the probe set with the smallest test statistic was kept for each gene [59], about 7,500 genes remained. (iii) Filtering out genes with low fold changes (FCs): Genes with log2(FC) < 0.67 × standard deviation (SD) away from the group mean (i.e., between the first and the third quartile assuming that log2(FC) follows a normal distribution) were removed. After these sequential steps of filtering, about 3,500 genes were left for downstream statistical analyses for each group. A schematic diagram of the detailed data mining and analysis plan is shown in Figure 3.

Figure 3
figure 3

Schematic diagram of study workflow , including probe set filtering steps and statistical test strategies for detecting significant single genes and pathways. The probe intensities measured in 209 hybridized Affymetrix HG-U133 plus 2.0 arrays were normalized by Robust Multichip Average followed by a baseline correction step. Probes marked ‘Presence’ in fewer than four arrays in each group (because for Week 12 placebo group, only two positive responders were included, probes with two valid measurements were kept) were removed. Probes corresponding to control or less well-defined genes, and duplicated probes were removed. Genes with low FCs; i.e., within 1 standard deviation (denoted by σ) for a total of L (~7500) genes also were removed, as most of them were not likely to be differentially expressed to a statistically significant extent. The remaining genes were tested by the ordinary Student’s t-test, and genes with P values < 0.05 were used for pathway analysis. In total, 3698, 3532, 3328, and 3405 genes were tested for the Week 8 TPM, Week 8 placebo, Week 12 TPM, and Week 12 placebo groups, respectively.

Statistical analysis to identify differentially expressed genes and pathways

After data quality checking, pre-processing, normalization, and probe set filtering, we analyzed the microarray data at both the single-gene level (where one seeks to determine whether each gene is expressed differently under different conditions) and the pathway level (where one intends to determine if a biological pathway shows a different expression pattern under different conditions). Considering the individual variations at the baseline, we normalized each individual’s Week 8 and Week 12 expression values by the corresponding baseline values prior to the identification of differentially expressed genes and biological pathways.

Single-gene analysis

The primary goal of this step is to detect those genes with significantly different expressions in two comparison groups that cannot be ascribed to chance or natural variability [60]. The ordinary Student’s t-test, implemented by MATLAB (MathWorks, Natick, MA), was employed for testing differential expressions in a gene-by-gene manner. To correct for multiple testing, both Bonferroni correction and false discovery rate (FDR); i.e., the expected proportion of falsely rejected null hypotheses among the rejected hypotheses, which was estimated by the Benjamini-Hochberg (BH) procedure [61], were applied.

Pathway analysis

Because gene expression is a well-coordinated system, expressions of different genes generally are not independent. Pathway analysis can reduce the number of hypotheses to a more manageable number that directly addresses questions of biological interest. During the past few years, various bioinformatics tools have been developed for pathway analysis, although none has gained widespread acceptance [60]. Therefore, in the current study, significantly enriched pathways of differentially expressed genes were detected using the following bioinformatics tools:

Ingenuity Pathway Analysis (IPA) (http://www.ingenuity.com/)

The IPA is a web-based bioinformatics tool [62]. A given set of input genes was associated with molecular networks based on their connectivities in the Ingenuity Pathways Knowledge Base. Fisher’s exact test was used to determine the probability that each biological function assigned to that data set was attributable to chance alone [63].

Onto-Tools Pathway-Express (http://vortex.cs.wayne.edu/projects.htm)

The Onto-Tools Pathway-Express [64],[65] implements an innovative “Impact Factor Analysis” based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Distinct from either “Over-Representation Analysis” (ORA) or “Gene Set Enrichment Analysis” (GSEA), Onto-Tools Pathway-Express uses a systems biology approach to identify pathways that are significantly impacted in any condition monitored by high-throughput gene expression technology. This new “Impact Factor Analysis” not only incorporates the classical probabilistic component but also includes important biological factors that are not captured by the existing techniques; e.g., the magnitude of the expression changes of each gene, the position of the differentially expressed genes on given pathways, the topology of the pathway that describes how genes interact, and the type of signaling interactions between them [64]. Based on a given set of input genes, for each pathway detected, a perturbation factor gamma P value and a corresponding FDR were calculated, taking into consideration the normalized FC of the gene and the number of genes upstream of its position in the pathway. Because IPA and Onto-Tools Pathway-Express have applied distinct statistical algorithms based on independent knowledge databases, these two bioinformatics tools are complementary, and thus their results are combined.

Additional files

References

  1. Yang MH, Jung MS, Lee MJ, Yoo KH, Yook YJ, Park EY, Choi SH, Suh YJ, Kim KW, Park JH: Gene expression profiling of the rewarding effect caused by methamphetamine in the mesolimbic dopamine system. Mol Cells. 2008, 26 (2): 121-130.

    CAS  PubMed  Google Scholar 

  2. Elkashef A, Vocci F, Hanson G, White J, Wickes W, Tiihonen J: Pharmacotherapy of methamphetamine addiction: an update. Subst Abus. 2008, 29 (3): 31-49. 10.1080/08897070802218554.

    Article  PubMed  PubMed Central  Google Scholar 

  3. SAMHSA: Results from the 2007 National Survey on Drug Use and Health: National Findings. Office of Applied Studies, NSDUH Series H-34, DHHS Publication No. SMA 08–4343. Rockville, MD: 2008.

  4. Akiyama K, Kanzaki A, Tsuchida K, Ujike H: Methamphetamine-induced behavioral sensitization and its implications for relapse of schizophrenia. Schizophr Res. 1994, 12 (3): 251-257. 10.1016/0920-9964(94)90035-3.

    Article  CAS  PubMed  Google Scholar 

  5. Anglin MD, Burke C, Perrochet B, Stamper E, Dawud-Noursi S: History of the methamphetamine problem. J Psychoactive Drugs. 2000, 32 (2): 137-141. 10.1080/02791072.2000.10400221.

    Article  CAS  PubMed  Google Scholar 

  6. Bisagno V, Ferguson D, Luine VN: Short toxic methamphetamine schedule impairs object recognition task in male rats. Brain Res. 2002, 940 (1–2): 95-101. 10.1016/S0006-8993(02)02599-4.

    Article  CAS  PubMed  Google Scholar 

  7. Nordahl TE, Salo R, Leamon M: Neuropsychological effects of chronic methamphetamine use on neurotransmitters and cognition: a review. J Neuropsychiatry Clin Neurosci. 2003, 15 (3): 317-325. 10.1176/jnp.15.3.317.

    Article  CAS  PubMed  Google Scholar 

  8. Schroder N, O’Dell SJ, Marshall JF: Neurotoxic methamphetamine regimen severely impairs recognition memory in rats. Synapse. 2003, 49 (2): 89-96. 10.1002/syn.10210.

    Article  PubMed  Google Scholar 

  9. Okochi T, Kishi T, Ikeda M, Kitajima T, Kinoshita Y, Kawashima K, Okumura T, Tsunoka T, Inada T, Yamada M, Uchimura N, Iyo M, Sora I, Ozaki N, Ujike H, Iwata N: Genetic association analysis of NRG1 with methamphetamine-induced psychosis in a Japanese population. Prog Neuropsychopharmacol Biol Psychiatry. 2009, 33 (5): 903-905. 10.1016/j.pnpbp.2009.04.016.

    Article  CAS  PubMed  Google Scholar 

  10. Davidson C, Gow AJ, Lee TH, Ellinwood EH: Methamphetamine neurotoxicity: necrotic and apoptotic mechanisms and relevance to human abuse and treatment. Brain Res Brain Res Rev. 2001, 36 (1): 1-22. 10.1016/S0165-0173(01)00054-6.

    Article  CAS  PubMed  Google Scholar 

  11. Ricaurte GA, Guillery RW, Seiden LS, Schuster CR, Moore RY: Dopamine nerve terminal degeneration produced by high doses of methylamphetamine in the rat brain. Brain Res. 1982, 235 (1): 93-103. 10.1016/0006-8993(82)90198-6.

    Article  CAS  PubMed  Google Scholar 

  12. Rocher C, Gardier AM: Effects of repeated systemic administration of d-Fenfluramine on serotonin and glutamate release in rat ventral hippocampus: comparison with methamphetamine using in vivo microdialysis. Naunyn Schmiedebergs Arch Pharmacol. 2001, 363 (4): 422-428. 10.1007/s002100000381.

    Article  CAS  PubMed  Google Scholar 

  13. Spina MB, Cohen G: Dopamine turnover and glutathione oxidation: implications for Parkinson disease. Proc Natl Acad Sci U S A. 1989, 86 (4): 1398-1400. 10.1073/pnas.86.4.1398.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tata DA, Yamamoto BK: Interactions between methamphetamine and environmental stress: role of oxidative stress, glutamate and mitochondrial dysfunction. Addiction. 2007, 102 (Suppl 1): 49-60. 10.1111/j.1360-0443.2007.01770.x.

    Article  PubMed  Google Scholar 

  15. Yamamoto BK, Zhu W: The effects of methamphetamine on the production of free radicals and oxidative stress. J Pharmacol Exp Ther. 1998, 287 (1): 107-114.

    CAS  PubMed  Google Scholar 

  16. Zhang X, Lee TH, Xiong X, Chen Q, Davidson C, Wetsel WC, Ellinwood EH: Methamphetamine induces long-term changes in GABAA receptor alpha2 subunit and GAD67 expression. Biochem Biophys Res Commun. 2006, 351 (1): 300-305. 10.1016/j.bbrc.2006.10.046.

    Article  CAS  PubMed  Google Scholar 

  17. Johnson BA, Ait-Daoud N, Elkashef AM, Smith EV, Kahn R, Vocci F, Li SH, Bloch DA: A preliminary randomized, double-blind, placebo-controlled study of the safety and efficacy of ondansetron in the treatment of methamphetamine dependence. Int J Neuropsychopharmacol. 2008, 11 (1): 1-14. 10.1017/S1461145707007778.

    Article  CAS  PubMed  Google Scholar 

  18. Maryanoff BE, Nortey SO, Gardocki JF, Shank RP, Dodgson SP: Anticonvulsant O-alkyl sulfamates. 2,3:4,5-Bis-O-(1-methylethylidene)-beta-D-fructopyranose sulfamate and related compounds. J Med Chem. 1987, 30 (5): 880-887. 10.1021/jm00388a023.

    Article  CAS  PubMed  Google Scholar 

  19. Johnson BA, Ait-Daoud N, Bowden CL, DiClemente CC, Roache JD, Lawson K, Javors MA, Ma JZ: Oral topiramate for treatment of alcohol dependence: a randomised controlled trial. Lancet. 2003, 361 (9370): 1677-1685. 10.1016/S0140-6736(03)13370-3.

    Article  CAS  PubMed  Google Scholar 

  20. Johnson BA, Ait-Daoud N, Akhtar FZ, Javors MA: Use of oral topiramate to promote smoking abstinence among alcohol-dependent smokers: a randomized controlled trial. Arch Intern Med. 2005, 165 (14): 1600-1605. 10.1001/archinte.165.14.1600.

    Article  CAS  PubMed  Google Scholar 

  21. Kampman KM, Pettinati H, Lynch KG, Dackis C, Sparkman T, Weigley C, O’Brien CP: A pilot trial of topiramate for the treatment of cocaine dependence. Drug Alcohol Depend. 2004, 75 (3): 233-240. 10.1016/j.drugalcdep.2004.03.008.

    Article  CAS  PubMed  Google Scholar 

  22. Johnson BA, Roache JD, Ait-Daoud N, Wells LT, Wallace CL, Dawes MA, Liu L, Wang XQ: Effects of topiramate on methamphetamine-induced changes in attentional and perceptual-motor skills of cognition in recently abstinent methamphetamine-dependent individuals. Prog Neuropsychopharmacol Biol Psychiatry. 2007, 31 (1): 123-130. 10.1016/j.pnpbp.2006.08.002.

    Article  CAS  PubMed  Google Scholar 

  23. Johnson BA, Roache JD, Ait-Daoud N, Wells LT, Wallace CL, Dawes MA, Liu L, Wang XQ: Effects of acute topiramate dosing on methamphetamine-induced subjective mood. Int J Neuropsychopharmacol. 2007, 10 (1): 85-98. 10.1017/S1461145705006401.

    Article  CAS  PubMed  Google Scholar 

  24. Johnson BA, Wells LT, Roache JD, Wallace CL, Ait-Daoud N, Dawes MA, Liu L, Wang XQ, Javors MA: Kinetic and cardiovascular effects of acute topiramate dosing among non-treatment-seeking, methamphetamine-dependent individuals. Prog Neuropsychopharmacol Biol Psychiatry. 2007, 31 (2): 455-461. 10.1016/j.pnpbp.2006.11.011.

    Article  CAS  PubMed  Google Scholar 

  25. Zullino DF, Benguettat D, Khazaal Y: Improvement of cognitive performance by topiramate: Blockage of automatic processes may be the underlying mechanism. Prog Neuropsychopharmacol Biol Psychiatry. 2007, 31 (3): 787-10.1016/j.pnpbp.2007.01.010.

    Article  CAS  PubMed  Google Scholar 

  26. Elkashef A, Kahn R, Yu E, Iturriaga E, Li SH, Anderson A, Chiang N, Ait-Daoud N, Weiss D, McSherry F, Serpi T, Rawson R, Hrymoc M, Weis D, McCann M, Pham T, Stock C, Dickinson R, Campbell J, Gorodetzky C, Haning W, Carlton B, Mawhinney J, Li MD, Johnson BA: Topiramate for the treatment of methamphetamine addiction: a multi-center placebo-controlled trial. Addiction. 2012, 107 (7): 1297-1306. 10.1111/j.1360-0443.2011.03771.x.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Mizoguchi H, Yamada K, Mizuno M, Mizuno T, Nitta A, Noda Y, Nabeshima T: Regulations of methamphetamine reward by extracellular signal-regulated kinase 1/2/ets-like gene-1 signaling pathway via the activation of dopamine receptors. Mol Pharmacol. 2004, 65 (5): 1293-1301. 10.1124/mol.65.5.1293.

    Article  CAS  PubMed  Google Scholar 

  28. Kauer JA, Malenka RC: Synaptic plasticity and addiction. Nat Rev Neurosci. 2007, 8 (11): 844-858. 10.1038/nrn2234.

    Article  CAS  PubMed  Google Scholar 

  29. Narita M, Miyatake M, Shibasaki M, Tsuda M, Koizumi S, Yajima Y, Inoue K, Suzuki T: Long-lasting change in brain dynamics induced by methamphetamine: enhancement of protein kinase C-dependent astrocytic response and behavioral sensitization. J Neurochem. 2005, 93 (6): 1383-1392. 10.1111/j.1471-4159.2005.03097.x.

    Article  CAS  PubMed  Google Scholar 

  30. Raffa RB, Finno KE, Tallarida CS, Rawls SM: Topiramate-antagonism of L-glutamate-induced paroxysms in planarians. Eur J Pharmacol. 2010, 649 (1–3): 150-153. 10.1016/j.ejphar.2010.09.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rawls SM, Thomas T, Adeola M, Patil T, Raymondi N, Poles A, Loo M, Raffa RB: Topiramate antagonizes NMDA- and AMPA-induced seizure-like activity in planarians. Pharmacol Biochem Behav. 2009, 93 (4): 363-367. 10.1016/j.pbb.2009.05.005.

    Article  CAS  PubMed  Google Scholar 

  32. Eun JW, Kwack SJ, Noh JH, Jung KH, Kim JK, Bae HJ, Xie H, Ryu JC, Ahn YM, Park WS, Lee JY, Rhee GS, Nam SW: Identification of post-generation effect of 3,4-methylenedioxymethamphetamine on the mouse brain by large-scale gene expression analysis. Toxicol Lett. 2010, 195 (1): 60-67. 10.1016/j.toxlet.2010.02.013.

    Article  CAS  PubMed  Google Scholar 

  33. Simoes PF, Silva AP, Pereira FC, Marques E, Grade S, Milhazes N, Borges F, Ribeiro CF, Macedo TR: Methamphetamine induces alterations on hippocampal NMDA and AMPA receptor subunit levels and impairs spatial working memory. Neuroscience. 2007, 150 (2): 433-441. 10.1016/j.neuroscience.2007.09.044.

    Article  CAS  PubMed  Google Scholar 

  34. Betancur C, Sakurai T, Buxbaum JD: The emerging role of synaptic cell-adhesion pathways in the pathogenesis of autism spectrum disorders. Trends Neurosci. 2009, 32 (7): 402-412. 10.1016/j.tins.2009.04.003.

    Article  CAS  PubMed  Google Scholar 

  35. Schluter OM, Xu W, Malenka RC: Alternative N-terminal domains of PSD-95 and SAP97 govern activity-dependent regulation of synaptic AMPA receptor function. Neuron. 2006, 51 (1): 99-111. 10.1016/j.neuron.2006.05.016.

    Article  CAS  PubMed  Google Scholar 

  36. Nakagawa T, Futai K, Lashuel HA, Lo I, Okamoto K, Walz T, Hayashi Y, Sheng M: Quaternary structure, protein dynamics, and synaptic function of SAP97 controlled by L27 domain interactions. Neuron. 2004, 44 (3): 453-467. 10.1016/j.neuron.2004.10.012.

    Article  CAS  PubMed  Google Scholar 

  37. Mehta S, Wu H, Garner CC, Marshall J: Molecular mechanisms regulating the differential association of kainate receptor subunits with SAP90/PSD-95 and SAP97. J Biol Chem. 2001, 276 (19): 16092-16099. 10.1074/jbc.M100643200.

    Article  CAS  PubMed  Google Scholar 

  38. Wang L, Piserchio A, Mierke DF: Structural characterization of the intermolecular interactions of synapse-associated protein-97 with the NR2B subunit of N-methyl-D-aspartate receptors. J Biol Chem. 2005, 280 (29): 26992-26996. 10.1074/jbc.M503555200.

    Article  CAS  PubMed  Google Scholar 

  39. Mauceri D, Gardoni F, Marcello E, Di Luca M: Dual role of CaMKII-dependent SAP97 phosphorylation in mediating trafficking and insertion of NMDA receptor subunit NR2A. J Neurochem. 2007, 100 (4): 1032-1046. 10.1111/j.1471-4159.2006.04267.x.

    Article  CAS  PubMed  Google Scholar 

  40. Suh YH, Terashima A, Petralia RS, Wenthold RJ, Isaac JT, Roche KW, Roche PA: A neuronal role for SNAP-23 in postsynaptic glutamate receptor trafficking. Nat Neurosci. 2010, 13 (3): 338-343. 10.1038/nn.2488.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wienecke J, Westerdahl AC, Hultborn H, Kiehn O, Ryge J: Global gene expression analysis of rodent motor neurons following spinal cord injury associates molecular mechanisms with development of postinjury spasticity. J Neurophysiol. 2010, 103 (2): 761-778. 10.1152/jn.00609.2009.

    Article  CAS  PubMed  Google Scholar 

  42. Hiraoka S, Kajii Y, Kuroda Y, Umino A, Nishikawa T: The development- and phencyclidine-regulated induction of synapse-associated protein-97 gene in the rat neocortex. Eur Neuropsychopharmacol. 2010, 20 (3): 176-186. 10.1016/j.euroneuro.2009.08.007.

    Article  CAS  PubMed  Google Scholar 

  43. Takaki M, Ujike H, Kodama M, Takehisa Y, Nakata K, Kuroda S: Two kinds of mitogen-activated protein kinase phosphatases, MKP-1 and MKP-3, are differentially activated by acute and chronic methamphetamine treatment in the rat brain. J Neurochem. 2001, 79 (3): 679-688. 10.1046/j.1471-4159.2001.00615.x.

    Article  CAS  PubMed  Google Scholar 

  44. Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, Steinman L: Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci U S A. 2010, 107 (6): 2580-2585. 10.1073/pnas.0915139107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sequeira A, Turecki G: Genome wide gene expression studies in mood disorders. OMICS. 2006, 10 (4): 444-454. 10.1089/omi.2006.10.444.

    Article  CAS  PubMed  Google Scholar 

  46. Churchill GA: Fundamentals of experimental design for cDNA microarrays. Nat Genet. 2002, 32 (Suppl 2): 490-495. 10.1038/ng1031.

    Article  CAS  PubMed  Google Scholar 

  47. van Leeuwen DM, van Agen E, Gottschalk RW, Vlietinck R, Gielen M, van Herwijnen MH, Maas LM, Kleinjans JC, van Delft JH: Cigarette smoke-induced differential gene expression in blood cells from monozygotic twin pairs. Carcinogenesis. 2007, 28 (3): 691-697. 10.1093/carcin/bgl199.

    Article  CAS  PubMed  Google Scholar 

  48. Lampe JW, Stepaniants SB, Mao M, Radich JP, Dai H, Linsley PS, Friend SH, Potter JD: Signatures of environmental exposures using peripheral leukocyte gene expression: tobacco smoke. Cancer Epidemiol Biomarkers Prev. 2004, 13 (3): 445-453.

    CAS  PubMed  Google Scholar 

  49. Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, Rosas HD, Hersch SM, Hogarth P, Bouzou B, Jensen RV, Krainc D: Genome-wide expression profiling of human blood reveals biomarkers for Huntington’s disease. Proc Natl Acad Sci U S A. 2005, 102 (31): 11023-11028. 10.1073/pnas.0504921102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Whitney AR, Diehn M, Popper SJ, Alizadeh AA, Boldrick JC, Relman DA, Brown PO: Individuality and variation in gene expression patterns in human blood. Proc Natl Acad Sci U S A. 2003, 100 (4): 1896-1901. 10.1073/pnas.252784499.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dumeaux V, Johansen J, Borresen-Dale AL, Lund E: Gene expression profiling of whole-blood samples from women exposed to hormone replacement therapy. Mol Cancer Ther. 2006, 5 (4): 868-876. 10.1158/1535-7163.MCT-05-0329.

    Article  CAS  PubMed  Google Scholar 

  52. Sullivan PF, Fan C, Perou CM: Evaluating the comparability of gene expression in blood and brain. Am J Med Genet B Neuropsychiatr Genet. 2006, 141B (3): 261-268. 10.1002/ajmg.b.30272.

    Article  PubMed  Google Scholar 

  53. Cai C, Langfelder P, Fuller TF, Oldham MC, Luo R, van den Berg LH, Ophoff RA, Horvath S: Is human blood a good surrogate for brain tissue in transcriptional studies?. BMC Genomics. 2010, 11: 589-10.1186/1471-2164-11-589.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Li C: Automating dChip: toward reproducible sharing of microarray data analysis. BMC Bioinformatics. 2008, 9: 231-10.1186/1471-2105-9-231.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP: Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003, 31 (4): e15-10.1093/nar/gng015.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Gautier L, Cope L, Bolstad BM, Irizarry RA: Affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 2004, 20 (3): 307-315. 10.1093/bioinformatics/btg405.

    Article  CAS  PubMed  Google Scholar 

  57. Calza S, Raffelsberger W, Ploner A, Sahel J, Leveillard T, Pawitan Y: Filtering genes to improve sensitivity in oligonucleotide microarray data analysis. Nucleic Acids Res. 2007, 35 (16): e102-10.1093/nar/gkm537.

    Article  PubMed  PubMed Central  Google Scholar 

  58. McClintick JN, Edenberg HJ: Effects of filtering by Present call on analysis of microarray experiments. BMC Bioinformatics. 2006, 7: 49-10.1186/1471-2105-7-49.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Rowe WB, Blalock EM, Chen KC, Kadish I, Wang D, Barrett JE, Thibault O, Porter NM, Rose GM, Landfield PW: Hippocampal expression analyses reveal selective association of immediate-early, neuroenergetic, and myelinogenic pathways with cognitive impairment in aged rats. J Neurosci. 2007, 27 (12): 3098-3110. 10.1523/JNEUROSCI.4163-06.2007.

    Article  CAS  PubMed  Google Scholar 

  60. Mutch DM, Berger A, Mansourian R, Rytz A, Roberts MA: The limit fold change model: a practical approach for selecting differentially expressed genes from microarray data. BMC Bioinformatics. 2002, 3: 17-10.1186/1471-2105-3-17.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc Series B. 1995, 57 (1): 289-300.

    Google Scholar 

  62. Nakatani N, Hattori E, Ohnishi T, Dean B, Iwayama Y, Matsumoto I, Kato T, Osumi N, Higuchi T, Niwa S, Yoshikawa T: Genome-wide expression analysis detects eight genes with robust alterations specific to bipolar I disorder: relevance to neuronal network perturbation. Hum Mol Genet. 2006, 15 (12): 1949-1962. 10.1093/hmg/ddl118.

    Article  CAS  PubMed  Google Scholar 

  63. Dozmorov MG, Kyker KD, Hauser PJ, Saban R, Buethe DD, Dozmorov I, Centola MB, Culkin DJ, Hurst RE: From microarray to biology: an integrated experimental, statistical and in silico analysis of how the extracellular matrix modulates the phenotype of cancer cells. BMC Bioinformatics. 2008, 9 (Suppl 9): S4-10.1186/1471-2105-9-S9-S4.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, Georgescu C, Romero R: A systems biology approach for pathway level analysis. Genome Res. 2007, 17 (10): 1537-1545. 10.1101/gr.6202607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Khatri P, Sellamuthu S, Malhotra P, Amin K, Done A, Draghici S: Recent additions and improvements to the Onto-Tools. Nucleic Acids Res. 2005, 33 (Web Server issue): W762-W765. 10.1093/nar/gki472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

The clinical trial data used for this study are registered in Clinicaltrials.gov with the identifier: NCT00345371. We are grateful for the invaluable contributions of clinical information and blood samples by all participants in this clinical trial, as well as the dedicated work of many research staff at the clinical sites. We are grateful to the National Institute on Drug Abuse for its generous support through the Department of Veterans Affairs Cooperative Studies Program (Interagency Agreement No. Y1-DA4006). In addition, statistical and bioinformatics analyses of expression data were in part supported by NIH grant DA-137873 to MDL and an NIH contract to IMC.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ming D Li.

Additional information

Competing interests

E Yu serves as a member of the scientific advisory board for US WorldMeds, LLC. MD Li serves as a board member for ADial Pharmaceuticals. B A Johnson has served as a consultant to Johnson & Johnson (Ortho-McNeil Janssen Scientific Affairs, LLC), Transcept Pharmaceuticals, Inc., D&A Pharma, Organon, ADial Pharmaceuticals, Psychological Education Publishing Company (PEPCo LLC), and Eli Lilly and Company. All other authors report no competing interests.

Authors’ contributions

NA-D, JC, WH, DJM, DW, MMC, CS, RK, EI, EY, AE, and BAJ participated in the clinical trial and contributed to both clinical and biological data collection. MDL, JW, TN, JZM, CS, DW, JS, and RM undertook the management, and statistical and bioinformatics analysis of data. MDL, JW, TN, and CS were involved in the interpretation of results and in writing of the manuscript. All authors contributed to and have approved the final manuscript.

Electronic supplementary material

12920_2014_65_MOESM1_ESM.pdf

Additional file 1: Figure S1.: Diagram of sample selection and grouping of responders and non-responders to TPM treatment and Placebo for Weeks 8 and 12. (PDF 53 KB)

12920_2014_65_MOESM2_ESM.doc

Additional file 2: Tables S1-4.: A detailed list of genes and pathways identified from the study at different treatment times. (DOC 1 MB)

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Authors’ original file for figure 3

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, M.D., Wang, J., Niu, T. et al. Transcriptome profiling and pathway analysis of genes expressed differentially in participants with or without a positive response to topiramate treatment for methamphetamine addiction. BMC Med Genomics 7, 65 (2014). https://doi.org/10.1186/s12920-014-0065-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12920-014-0065-x

Keywords