Abstract
Background
The behavioral consequences associated with addiction are thought to arise from drug-induced neuroadaptation. The mesolimbic system plays an important initial role in this process, and while the dopaminergic system specifically has been strongly interrogated, a complete understanding of the broad transcriptomic effects associated with cocaine use remains elusive.
Methods
Using next generation sequencing approaches, we performed a comprehensive evaluation of gene expression differences in the ventral tegmental area and nucleus accumbens of rhesus macaques that had self-administered cocaine for roughly 100 days and saline-yoked controls. During self-administration, the monkeys increased daily consumption of cocaine until almost the maximum number of injections were taken within the first 15 min of the one hour session for a total intake of 3 mg/kg/day.
Results
We confirm the centrality of dopaminergic differences in the ventral tegmental area, but in the nucleus accumbens we see the strongest evidence for an inflammatory response and large scale chromatin remodeling.
Conclusions
These findings suggest an expanded understanding of the pathology of cocaine addiction with the potential to lead to the development of alternative treatment strategies.
Keywords: mesolimbic pathway, cocaine, rhesus monkey, genetics, neuroinflammation
1. Introduction
Drug abuse is associated with a number of molecular and cellular effects on the brain including changes in neurocircuitry, gene expression, and epigenetic regulation. These changes are believed to be linked with the transition from substance use to abuse: compulsive drug seeking, loss of restraint, and negative affect (Koob and Volkow, 2010; Luscher and Malenka, 2011). The mesolimbic dopamine system, connecting the ventral tegmental area (VTA) and the nucleus accumbens (NAc), drives the salient effects of reward and has been consistently and repeatedly shown to be the primary site of action of drugs of abuse (Nestler, 2005).
There have been a number of studies in rodents and human post-mortem tissues that focus on gene expression differences associated with addiction both in the mesolimbic pathway as well as other areas of the brain (Zhou et al., 2014b). Microarray studies in rodents have identified differences in immediate-early genes (genes activated rapidly following extracellular stimulation) and dopaminergic pathways (Piechota et al., 2010; Yuferov et al., 2003; Zhang et al., 2005) in accordance with previous findings using Northern blots and immunohistochemistry (Hope et al., 1992). More recent studies using next generation sequencing technologies to study the effect of cocaine on the mouse NAc found differences from controls across multiple neurotransmitter systems including dopaminergic, cholinergic, glutamatergic, GABAergic systems as well as cadherin and Wnt signaling pathways (Eipper-Mains et al., 2013). This has been extended to non-coding RNAs with functional effects imputed using informatics approaches (Bu et al., 2012; Chen et al., 2013).
There have also been a number of microarray studies of gene expression on human cocaine abusers in the NAc (Albertson et al., 2004; Bannon et al., 2005) and midbrain dopaminergic regions (Tang et al., 2003). While confirming many of the dopaminergic findings from rodents, more widespread transcriptional changes were also identified. These findings have been suggested to reflect epigenetic reprogramming resulting from chronic drug exposure (Zhou et al., 2011). The differences observed between rodent and human expression studies may result from methodological differences in the duration of exposure to drug, acute compared to chronic usage (Zhou et al., 2014b). Indeed, transgenic mouse work has shown that histone acetylation is important for response to chronic, but not acute, cocaine exposure (Renthal et al., 2007).
One of the challenges in developing translational animal models is to recapitulate as closely as possible the most salient features of human behavior while maintaining precise experimental control. In addition to greater genetic and neuroanatomical similarities, nonhuman primate models in particular are valuable when modeling aspects of addiction because their consumption and patterns of drug taking so closely reflect that seen in human drug addiction (Platt and Rowlett, 2012). A rhesus macaque model of self-administration provides a unique opportunity to explore the molecular mechanisms involved in the entirety of the neurochemical systems and circuitry associated with the addictive processes (Weerts et al., 2007).
In the present study, we used a cocaine self-administration procedure in rhesus macaques over three months (approximately 100 consecutive days). After this exposure period, transcriptomic analysis was performed on the NAc and the VTA using next generation RNA-seq. This allows for an unbiased and holistic view of the differences between cocaine- and saline-treated animals in these regions and provides a simultaneous assessment of the two brain regions in long-term cocaine consumption.
2. Materials and Methods
2.1 Ethics statement
Animals were maintained in accordance with the guidelines of the Committee on Animals of Harvard Medical School and the Guide for the Care and Use of Laboratory Animals (8th edition, 2011). Research protocols were approved by the Harvard Medical School Institutional Animal Care and Use Committee.
2.2 Animals
Subjects were 10, experimentally naïve, male young adult (4–7 years) rhesus macaques (Macaca mulatta). All animals were raised in shared conditions with identical diet and husbandry; paired animals balanced for age and weight were randomly assigned to cocaine exposure or yoked saline. All animals were of Indian ancestry and unrelated to at least kinship coefficient < 0.02. Prior to the cocaine self-administration protocol, drug exposure was limited to periodic sedation (~1 injection/quarterly) with ketamine, a commonly used veterinary sedative, for preventative healthcare, isoflurane anesthesia for surgery, and post-operative antibiotics. All monkeys were housed individually and maintained on a 12-hr lights-on/12-hr lights-off cycle (lights on at 7:00 AM) with water available continuously. Monkeys received Teklad monkey diet, supplemented with fruits and vegetables, at least 1 hour after the end of the daily session, in quantities that allowed them to gain no more than 1 kg during the 100+ days of the study. Initial weights were 6–8 kg, with no significant changes noted over the course of the experiment.
Monkeys were prepared with a chronic indwelling venous catheter (polyvinyl chloride, i.d.: 0.64 mm; o.d.: 1.35 mm) according to previously described procedures (Platt et al., 2011). Monkeys were anesthetized initially with 10–20 mg/kg i.m. of ketamine. Throughout surgery, anesthesia was maintained by an isoflurane/oxygen mixture. Under aseptic conditions, a catheter was implanted in the internal jugular vein and passed to the level of the right atrium. The distal end of the catheter was passed subcutaneously and exited in the mid-scapular region. The external end of the catheter was fed through a fitted jacket and tether system (Lomir Biomedical, Toronto, Canada) and attached to a fluid swivel mounted to the animal’s cage. The catheters were flushed daily with heparinized saline (150–200 U/mL).
2.3 Self-administration and yoked saline control
Daily drug self-administration sessions occurred in each monkey’s home cage (MetalSmiths, Boston, MA). Monkeys were trained to self-administer cocaine (0.03 mg/kg/injection) under a 1-response, fixed-ratio schedule (FR 1) of i.v. drug injection. At the beginning of each session, a set of two white stimulus lights above a response lever was illuminated (Med Associates, St Albans, VT). Upon pressing the lever, the white lights were extinguished and a set of two red stimulus lights were illuminated for 1-s, coinciding with a 1-s infusion. Sessions were available for 1 h or 100 injections, whichever occurred first. This allowed for a maximum of 3 mg/kg cocaine per day, an amount producing relevant physiological effects in the rhesus macaque and corresponding to recreational doses in humans (Barnett et al., 1981). A single session occurred each day, 7 days/week, until a minimum of 100 sessions occurred for each monkey (achieved for 4 of 5 monkeys). A target cumulative dose of 300 mg/kg (i.e., 0.03 mg/kg/injection x daily number of injections x total number of sessions) was chosen a priori for terminating the study.
In order to determine the extent to which transcriptional differences were due to cocaine self-administration, a yoked design was employed for these studies. Each cocaine self-administration monkey was paired with an age- and weight-matched control monkey. The monkey’s surgeries were conducted on consecutive days, and the dyad was housed in the same room, although not directly beside the matched animal. The lever apparatus was made available to the yoked monkey, and the light conditions were the same: Two white stimulus lights above the lever were illuminated at the beginning of the session. When the cocaine self-administration monkey of the dyad pressed the lever, the white lights were extinguished and the red lights were illuminated, coinciding with a 1-s infusion of drug for the cocaine animal and saline for the yoked animal. For the yoked saline monkey, pressing the lever had no programmed consequences and thus the behavior was infrequent. The yoked saline monkey was euthanized on the same day as the matched cocaine monkey, but tissue collection occurred sequentially rather than simultaneously (the order of yoked saline vs. matched cocaine was counterbalanced). Therefore, the saline control animals experienced all conditions that were experienced by the cocaine self-administration monkeys with the exception of cocaine exposure.
2.4 RNA isolation and library preparation
All monkeys were euthanized approximately 24 h after the last session. Animals are sedated with ketamine (10–15 mg/kg, IM) followed by sodium pentobarbital (>75 mg/kg, IV) to effect. Standard gross pathology is then conducted and tissues removed. No more than three hours elapsed between euthanasia and freezing of samples. Brains were manually sliced into coronal sections with 2mm thickness from the frontal pole to the end of the midbrain. Tissues were stored at −80°C until all samples were collected. When all samples were available, two brain regions (NAc and VTA) were punched from brain blocks for use in molecular studies. The boundaries for the VTA were medial and slightly ventral to the substantia nigra compacta (SNC) and ventral and slightly lateral to the ventral medial nucleus of the hypothalamus (VMH) at the level of the oculomotor nerve (3n). The boundaries of the NAc were ventral and medial to the putamen and anterior commissure at the level of the optic chiasm and slightly dorsal to the anterior hypothalamic area (AHA). No attempt was made to dissociate shell and core subregions, opting instead for a more inclusive definition of the region of interest. All regions were identified and defined based on published literature (Paxinos et al., 2000). Samples were removed from the anterior to posterior limits for each animal and adjacent sections were combined to create a single regional sample for each animal. All samples were processed contemporaneously for RNA extraction using a standard Trizol protocol (Life Technology, Carlsbad, CA). RNA integrity was tested using an Agilent 2100 Bioanalyzer (Agilent Technologies Inc., Santa Clara, CA). All the samples found to have an RNA integrity number ≥ 8 were considered adequate and sufficient for further use (Schroeder et al., 2006).
2.5 μg of total RNA from each sample was used for transcriptomic analysis. The polyA mRNA was selected using an IntegenX Apollo 324 robot and associated PrepX-PolyA kit according to manufacturer’s protocols (IntegenX Inc., Pleasanton, CA). Library size and concentration was evaluated for quality using an Agilent D1K High Sensitivity DNA chip (Agilent Technologies Inc., Santa Clara, CA) followed by a SYBR qPCR assay performed using a Stratagene MxPro 3005P qPCR System. Duplicate measurements were carried out following previously established protocols (Meyer et al., 2008). Libraries were pooled, index tagged, and multiplexed 5 samples/lane, in equimolar amounts, and then denatured, clustered and sequenced on the Illumina HiSeq 2500 using a 50bp single end read protocol. Library preparation, and next generation sequencing was performed at the Biopolymers Facility, Department of Genetics, Harvard Medical School, Boston, MA.
2.5 Statistical and bioinformatic analysis
Cocaine self-administration was recorded as the number of injections per daily session. One animal (C2) had a catheter failure and only completed 84 days of cocaine self-administration; the remainder achieved 100 of cocaine self-administration. A linear-trend analysis was conducted on the number of injections/session across the 84 days for all monkeys and 100 days for the subset. Daily sessions were parsed into 4 bins of 15 min each and the number of injections that occurred in each bin was analyzed in order to assess within-session patterns of consumption. The injections/bin data were averaged for the first week (sessions 1–7), a mid-point (sessions 47–53), and final week (sessions 93–100). Sessions were grouped to minimize stochastic variability across individual days. An extra sum of squares F test was used following linear regression to detect a non-zero slope. The timing of self administration was analyzed with 2-way repeated measures ANOVA with Bonferroni correction for multiple comparisons.
Initial transcriptomic analysis was processed through DNAnexus (DNAnexus Inc., Mountain View, CA). All reads were initially aligned to the rhesus genome (MGSC Merged 1.0/rheMac2) and annotated using RefSeq annotations. Sample read depth was normalized using total count normalization and differentially expressed genes were identified using a Poisson log-linear model as implemented in the ‘PoissonSeq’ package for R.(Li et al., 2012) This methodology calculates a single gene significance value (p-value) as well as a false discovery rate corrected significance value (q-value).
A reanalysis of the data was performed following the publication of a new rhesus macaque genome (Mmul8.0.1) in early 2016. This analysis used the TopHat2 (Trapnell et al., 2012) suite of tools including ‘bowtie2’ for alignment, ‘cufflinks’ for transcript identification (again using the new RefSeq annotation), ‘cuffdiff’ to identify differentially expressed transcripts across conditions, and ‘cummeRbund’ for visualization. While this newer reference genome did improve the percent of mapped reads (roughly 10% greater mapping to the new genome) and marginally affect the significance values of the differential expression analysis, it did not change either the specific, or general, conclusions described herein. Heatmaps were generated using k-means clustering based on the Jensen-Shannon distance to group genes of similar expression profiles.
Data were analyzed using QIAGEN’s IngenuityR Pathway Analysis (IPAR, QIAGEN Redwood City, www.qiagen.com/ingenuity). IPA uses an extensive knowledge base to recapitulate biological networks and to associate biological function with genes in a regulated ontology. Overrepresented categories within the differentially expressed genes are identified using a right-tailed Fisher’s exact test with Benjamini-Hochberg correction for multiple testing. A q-value of 0.05 for these categories was used for further analysis.
3. Results
3.1 Cocaine self-administration
While there is some variability across animals (Supplemental Figure 11), when cocaine self-administration was averaged across animals, there was evidence of a gradual increase in self-administration over sessions (Figure 1A,1B). Linear trend analysis revealed a significant fit of these data (84 days (n=5) [F(1,82)=18.23, p<0.001]; 100 days (n=4) [F(1,98)= 25.06, p<0.001]) with a positive slope, although the relationship of session and number of injections/hour was relatively weak (r2= 0.18 or 0.20 respectively). A relatively robust increase in self-administration was seen when the session was separated into 15-min bins (Figure 1C). In general, more self-administration was observed in the first 15 minutes than the other bins, with the mean number of injections/session increasing significantly over sessions for the first bin (1–7 vs. 47–53 [t(48)=6.307, p<0.001]; 1–7 vs. 93–100: [t(48)=8.601, p <0.001]) but not the other bins. In fact, there is some evidence for a decrease in self-administration from later bins, coincident with the increase in injections earlier in the session. Although a possible reason for the lower numbers in the latter bins could have been the animals finishing the sessions early (i.e., completing all 100 injections prior to the end of the 60-min period), this was not observed. Further analyses of the time required to complete the sessions, either by self-administering all 100 injections or by “timing out” after 60 minutes, over the same 1-week time intervals revealed no significant effects (data not shown) with all sessions ending at or near the 60-min mark. While there is some inter-individual variability with regard to timing of injections, it is not significant and represents a much lesser effect than intra-individual across days. Generally, as animals progress there is a modest increase in total amount of cocaine self-administered across the entirety of the session with a more substantial shift to rapid consumption immediately upon availability.
Figure 1.
Grouped data is shown for 5 rhesus makes self-administering cocaine. Average number of injections/session are shown over the course of the 100 days (n=4, A) or 84 days (n=5, B). A significant (p < 0.05) linear escalation is observed. (C) Timing of self-administered injections (grouped in 15 minute bins) are shown for the first and last weeks of study and the midpoint week. The preponderance of injections in each case occur in the first 15 minute block. It is during this time block that a significant change (p < 0.05) is observed over the course of the study.
3.2 mRNA expression profiling
At the end of the behavioral study, animals were sacrificed and brains were removed and dissected, keeping target regions intact. The VTA and NAc were isolated for subsequent transcriptomic analysis using standard single-end RNA-seq technologies as described above. An average of 30.5 million reads (1,350 Mbases) were sequenced from each sample (Supplemental Table 1,2). Of these, an average of 54% (ranging from 4.5 million reads to 35.8 million reads/sample) were successfully mapped to the original, rheMac2, rhesus genome. A subsequent realignment to the newly released Mmul8.0.1 genome raised the average mapping to 63%. The RefSeq annotation of the rhesus genome was used for subsequent analyses though Ensembl annotations did not produce significantly different results.
We used a false discovery rate (FDR, q<0.05) that was conservative (Storey and Tibshirani, 2003). The number of genes identified in this manner, both before and after correction for multiple testing, was roughly equivalent to a similar study focused on human postmortem hippocampus following chronic cocaine exposure (80 genes differentially expressed at q<0.05) (Zhou et al., 2011). Overall a smaller number of genes were found to be differentially expressed (q <0.05) in the VTA (53; Table 1) compared to the NAc (328; Table 2). While there were more genes upregulated than downregulated in the VTA were more similar (66% and 34% respectively, Figure 2A), in the NAc this trend was much more exaggerated (96% to 4%, Figure 2B). The relative magnitude of differences in gene expression overall were also greater in the NAc compared to the VTA. Moreover, while genes showing effects in the VTA largely were associated with nervous system development and function (p < 0.0001), this pattern was entirely absent in the NAc. Instead, the most enriched categories of genes were those associated with inflammatory response (p < 0.001) notably including macrophage recruitment (p < 0.0005) and activation (p < 0.005) and leukocyte apoptosis (p < 0.0005).
Table 1.
Genes differentially expressed (q < 0.05) in the ventral tegmental area of rhesus macaques with long term cocaine self-administration compared to yoked controls.
P-value | FDR (q-value) | Log Ratio | Symbol | Entrez Gene Name |
---|---|---|---|---|
0.0000 | 0.0000 | 1.918 | CRYM | crystallin, mu |
0.0000 | 0.0000 | −1.761 | C1QL1 | complement component 1, q subcomponent-like 1 |
0.0000 | 0.0000 | −2.349 | CHRNA2 | cholinergic receptor, nicotinic, alpha 2 (neuronal) |
0.0000 | 0.0157 | 1.387 | DIRAS3 | DIRAS family, GTP-binding RAS-like 3 |
0.0000 | 0.0157 | −1.704 | FOXA2 | forkhead box A2 |
0.0000 | 0.0200 | 1.436 | CBLN4 | cerebellin 4 precursor |
0.0000 | 0.0200 | −2.934 | SLC6A3 | solute carrier family 6 (neurotransmitter transporter), member 3 |
0.0000 | 0.0200 | −2.099 | NKX6-1 | NK6 homeobox 1 |
0.0000 | 0.0200 | 1.109 | TPD52L1 | tumor protein D52-like 1 |
0.0000 | 0.0200 | −1.343 | CA8 | carbonic anhydrase VIII |
0.0000 | 0.0221 | 4.748 | PTH2 | parathyroid hormone 2 |
0.0001 | 0.0221 | −1.144 | SLC17A8 | solute carrier family 17 (vesicular glutamate transporter), member 8 |
0.0001 | 0.0221 | 1.247 | LRRC23 | leucine rich repeat containing 23 |
0.0001 | 0.0221 | 1.225 | DNAH12 | dynein, axonemal, heavy chain 12 |
0.0001 | 0.0221 | 1.28 | ARX | aristaless related homeobox |
0.0001 | 0.0221 | 1.158 | DYDC2 | DPY30 domain containing 2 |
0.0001 | 0.0221 | 2.229 | SLC18A3 | solute carrier family 18 (vesicular acetylcholine transporter), member 3 |
0.0001 | 0.0221 | 1.157 | DNAH5 | dynein, axonemal, heavy chain 5 |
0.0001 | 0.0221 | 2.599 | TLL1 | tolloid-like 1 |
0.0002 | 0.0221 | 1.537 | CALCB | calcitonin-related polypeptide beta |
0.0002 | 0.0221 | −1.437 | TH | tyrosine hydroxylase |
0.0002 | 0.0221 | −1.231 | FOXA1 | forkhead box A1 |
0.0003 | 0.0221 | 1.468 | CXorf30 | chromosome X open reading frame 30 |
0.0003 | 0.0221 | −1.423 | ASAH2 | N-acylsphingosine amidohydrolase (non-lysosomal ceramidase) 2 |
0.0003 | 0.0221 | 1.367 | AKAP14 | A kinase (PRKA) anchor protein 14 |
0.0003 | 0.0221 | 1.426 | AK9 | adenylate kinase 9 |
0.0003 | 0.0221 | 1.242 | CCDC60 | coiled-coil domain containing 60 |
0.0003 | 0.0221 | 1.318 | VSIG8 | V-set and immunoglobulin domain containing 8 |
0.0004 | 0.0221 | 1.483 | NAALADL1 | N-acetylated alpha-linked acidic dipeptidase-like 1 |
0.0004 | 0.0221 | −1.574 | DDC | dopa decarboxylase (aromatic L-amino acid decarboxylase) |
0.0004 | 0.0221 | 1.169 | IQGAP2 | IQ motif containing GTPase activating protein 2 |
0.0004 | 0.0221 | 1.151 | KRT18 | keratin 18 |
0.0004 | 0.0221 | 1.998 | SNRPD2 | small nuclear ribonucleoprotein D2 polypeptide 16.5kDa |
0.0004 | 0.0222 | 1.171 | AGR3 | anterior gradient 3 |
0.0004 | 0.0222 | −1.219 | EBF3 | early B-cell factor 3 |
0.0005 | 0.0227 | 1.358 | PPP1R32 | protein phosphatase 1, regulatory subunit 32 |
0.0005 | 0.0227 | 1.23 | C11orf70 | chromosome 11 open reading frame 70 |
0.0005 | 0.0227 | 1.101 | MYB | v-myb avian myeloblastosis viral oncogene homolog |
0.0006 | 0.0230 | 1.334 | CTXN3 | cortexin 3 |
0.0006 | 0.0230 | −1.376 | FOXD2 | forkhead box D2 |
0.0006 | 0.0231 | −1.294 | DMRTA2 | DMRT-like family A2 |
0.0006 | 0.0232 | −1.303 | MYOC | myocilin, trabecular meshwork inducible glucocorticoid response |
0.0006 | 0.0236 | −1.498 | TWIST1 | twist family bHLH transcription factor 1 |
0.0007 | 0.0238 | 1.723 | CDH9 | cadherin 9, type 2 (T1-cadherin) |
0.0008 | 0.0256 | 1.556 | GPC3 | glypican 3 |
0.0009 | 0.0270 | 1.389 | TRHDE | thyrotropin-releasing hormone degrading enzyme |
0.0010 | 0.0288 | 1.103 | MARCH10 | membrane-associated ring finger (C3HC4) 10, E3 ubiquitin protein ligase |
0.0011 | 0.0293 | 1.18 | ANKFN1 | ankyrin-repeat and fibronectin type III domain containing 1 |
0.0012 | 0.0297 | 1.236 | KCNB2 | potassium voltage-gated channel, Shab-related subfamily, member 2 |
0.0012 | 0.0297 | 1.107 | LMO1 | LIM domain only 1 (rhombotin 1) |
0.0012 | 0.0297 | −1.635 | T | T, brachyury homolog (mouse) |
0.0024 | 0.0398 | 1.456 | OSBPL3 | oxysterol binding protein-like 3 |
0.0024 | 0.0404 | −1.332 | TK1 | thymidine kinase 1, soluble |
Supplementary material can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:...
Table 2.
Genes differentially expressed (q < 0.05) in the nucleus accumbens of rhesus macaques with long term cocaine self-administration compared to yoked controls.
p-value | FDR (q-value) | Log Ratio | Symbol | Entrez Gene Name |
---|---|---|---|---|
0.0000 | 0.0000 | 1.249 | ATP2B4 | ATPase, Ca++ transporting, plasma membrane 4 |
0.0000 | 0.0000 | 7.353 | LECT2 | leukocyte cell-derived chemotaxin 2 |
0.0000 | 0.0016 | 1.271 | C15orf26 | chromosome 15 open reading frame 26 |
0.0000 | 0.0016 | 2.637 | C3orf70 | chromosome 3 open reading frame 70 |
0.0000 | 0.0016 | 1.604 | C6orf57 | chromosome 6 open reading frame 57 |
0.0000 | 0.0016 | 3.098 | FAM129C | family with sequence similarity 129, member C |
0.0000 | 0.0016 | 2.161 | FAM76A | family with sequence similarity 76, member A |
0.0000 | 0.0016 | 2.24 | H3F3A/H3F3B | H3 histone, family 3A |
0.0000 | 0.0016 | 4.335 | HBM | hemoglobin, mu |
0.0000 | 0.0016 | 3.716 | LILRA4 | leukocyte immunoglobulin-like receptor, subfamily A (with TM domain), member 4 |
0.0000 | 0.0016 | 4.799 | RAG2 | recombination activating gene 2 |
0.0000 | 0.0016 | 5.25 | RPL21 | ribosomal protein L21 |
0.0000 | 0.0016 | 2.271 | RPL36 | ribosomal protein L36 |
0.0000 | 0.0016 | 2.255 | SFRP4 | secreted frizzled-related protein 4 |
0.0000 | 0.0033 | 1.194 | STX17 | syntaxin 17 |
0.0000 | 0.0035 | 3.561 | REP15 | RAB15 effector protein |
0.0000 | 0.0035 | 1.849 | RPL36A | ribosomal protein L36a |
0.0000 | 0.0035 | 1.994 | SLC2A4 | solute carrier family 2 (facilitated glucose transporter), member 4 |
0.0000 | 0.0035 | 5.53 | SLPI | secretory leukocyte peptidase inhibitor |
0.0000 | 0.0035 | 1.94 | ZBED4 | zinc finger, BED-type containing 4 |
0.0000 | 0.0037 | 4.155 | RPL37 | ribosomal protein L37 |
0.0000 | 0.0039 | 4.218 | ADIG | adipogenin |
0.0000 | 0.0039 | 2.311 | RAB22A | RAB22A, member RAS oncogene family |
0.0000 | 0.0041 | 2.808 | ATP5L | ATP synthase, H+ transporting, mitochondrial Fo complex, subunit G |
0.0000 | 0.0041 | 1.709 | MYC | v-myc avian myelocytomatosis viral oncogene homolog |
0.0000 | 0.0041 | 1.523 | PCDHB1 | protocadherin beta 1 |
0.0000 | 0.0041 | 2.413 | SPA17 | sperm autoantigenic protein 17 |
0.0000 | 0.0041 | 3.033 | UTS2 | urotensin 2 |
0.0000 | 0.0042 | 3.482 | SLC22A8 | solute carrier family 22 (organic anion transporter), member 8 |
0.0000 | 0.0044 | 5.432 | CLDN16 | claudin 16 |
0.0000 | 0.0044 | 2.153 | ELK4 | ELK4, ETS-domain protein (SRF accessory protein 1) |
0.0000 | 0.0046 | 2.133 | IRF6 | interferon regulatory factor 6 |
0.0000 | 0.0046 | 1.564 | WNT4 | wingless-type MMTV integration site family, member 4 |
0.0000 | 0.0047 | 4.107 | DEFA5 | defensin, alpha 5, Paneth cell-specific |
0.0000 | 0.0047 | 2.192 | TLR10 | toll-like receptor 10 |
0.0000 | 0.0055 | 2.139 | FMN1 | formin 1 |
0.0000 | 0.0055 | 2.322 | RPL31 | ribosomal protein L31 |
0.0000 | 0.0055 | 3.147 | RPLP1 | ribosomal protein, large, P1 |
0.0000 | 0.0062 | 2.71 | DEFB119 | defensin, beta 119 |
0.0000 | 0.0062 | 3.177 | RPL10L | ribosomal protein L10-like |
0.0000 | 0.0065 | 3.804 | DSG4 | desmoglein 4 |
0.0001 | 0.0070 | 2.32 | GCNT3 | glucosaminyl (N-acetyl) transferase 3, mucin type |
0.0001 | 0.0070 | 1.908 | MAP1LC3B2 | microtubule-associated protein 1 light chain 3 beta 2 |
0.0001 | 0.0070 | 3.009 | MYCT1 | myc target 1 |
0.0001 | 0.0070 | 4.163 | OR1M1 | olfactory receptor, family 1, subfamily M, member 1 |
0.0000 | 0.0070 | 4.609 | OR8B8 | olfactory receptor, family 8, subfamily B, member 8 |
0.0000 | 0.0070 | 2.166 | RPL17 | ribosomal protein L17 |
0.0001 | 0.0070 | 2.342 | RPS27A | ribosomal protein S27a |
0.0001 | 0.0070 | 1.961 | TMEM45B | transmembrane protein 45B |
0.0001 | 0.0075 | 2.919 | CYP3A43 | cytochrome P450, family 3, subfamily A, polypeptide 43 |
0.0001 | 0.0075 | 3.645 | DEFB121 | defensin, beta 121 |
0.0001 | 0.0075 | 1.608 | KIAA1958 | KIAA1958 |
0.0001 | 0.0075 | 2.026 | LDHAL6B | lactate dehydrogenase A-like 6B |
0.0001 | 0.0075 | 1.152 | UQCRH | ubiquinol-cytochrome c reductase hinge protein |
0.0001 | 0.0075 | 1.453 | FAM196B | family with sequence similarity 196, member B |
0.0001 | 0.0076 | 3.561 | RPL12 | ribosomal protein L12 |
0.0001 | 0.0079 | 1.74 | GABRB2 | gamma-aminobutyric acid (GABA) A receptor, beta 2 |
0.0001 | 0.0079 | 2.94 | YDJC | YdjC homolog (bacterial) |
0.0001 | 0.0081 | 1.383 | CLN8 | ceroid-lipofuscinosis, neuronal 8 (epilepsy, progressive with mental retardation) |
0.0001 | 0.0081 | 1.764 | PSD3 | pleckstrin and Sec7 domain containing 3 |
0.0001 | 0.0081 | 1.374 | TRAPPC3L | trafficking protein particle complex 3-like |
0.0001 | 0.0081 | 1.531 | ZMAT3 | zinc finger, matrin-type 3 |
0.0001 | 0.0082 | 1.748 | PGAP1 | post-GPI attachment to proteins 1 |
0.0001 | 0.0087 | 3.735 | CLEC9A | C-type lectin domain family 9, member A |
0.0001 | 0.0087 | 1.236 | CTU1 | cytosolic thiouridylase subunit 1 |
0.0001 | 0.0087 | 3.723 | CXCL8 | chemokine (C-X-C motif) ligand 8 |
0.0001 | 0.0087 | 2.625 | KBTBD13 | kelch repeat and BTB (POZ) domain containing 13 |
0.0001 | 0.0087 | 3.448 | LCN8 | lipocalin 8 |
0.0001 | 0.0087 | 3.668 | NPSR1 | neuropeptide S receptor 1 |
0.0001 | 0.0087 | 1.316 | TMEM156 | transmembrane protein 156 |
0.0001 | 0.0088 | 2.365 | HMGN2 | high mobility group nucleosomal binding domain 2 |
0.0001 | 0.0089 | 1.726 | CATSPER3 | cation channel, sperm associated 3 |
0.0001 | 0.0089 | 1.985 | ELSPBP1 | epididymal sperm binding protein 1 |
0.0001 | 0.0089 | 1.728 | PCP2 | Purkinje cell protein 2 |
0.0001 | 0.0089 | 3.16 | TNFSF15 | tumor necrosis factor (ligand) superfamily, member 15 |
0.0001 | 0.0089 | 1.888 | ZNF483 | zinc finger protein 483 |
0.0001 | 0.0090 | 2.422 | USP6 | ubiquitin specific peptidase 6 |
0.0001 | 0.0090 | 1.369 | SCD | stearoyl-CoA desaturase (delta-9-desaturase) |
0.0001 | 0.0092 | 2.676 | C10orf82 | chromosome 10 open reading frame 82 |
0.0002 | 0.0094 | 1.507 | HMX1 | H6 family homeobox 1 |
0.0002 | 0.0100 | 4.809 | IRF2BP2 | interferon regulatory factor 2 binding protein 2 |
0.0002 | 0.0100 | 2.439 | RC3H1 | ring finger and CCCH-type domains 1 |
0.0002 | 0.0103 | 1.355 | NDUFA1 | NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 1, 7.5kDa |
0.0002 | 0.0105 | 2.368 | CLRN1 | clarin 1 |
0.0002 | 0.0105 | 3.849 | CTLA4 | cytotoxic T-lymphocyte-associated protein 4 |
0.0002 | 0.0105 | 2.812 | CXCL1 | chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha) |
0.0002 | 0.0105 | 2.021 | GPR160 | G protein-coupled receptor 160 |
0.0002 | 0.0105 | 2.831 | GTSF1 | gametocyte specific factor 1 |
0.0002 | 0.0105 | 2.122 | LOC102723532/OR4N4 | olfactory receptor, family 4, subfamily N, member 4 |
0.0002 | 0.0105 | 1.618 | MALAT1 | metastasis associated lung adenocarcinoma transcript 1 (non-protein coding) |
0.0002 | 0.0105 | 2.169 | MCHR2 | melanin-concentrating hormone receptor 2 |
0.0002 | 0.0105 | 2.383 | PPP3R2 | protein phosphatase 3, regulatory subunit B, beta |
0.0002 | 0.0105 | 3.612 | PRR30 | proline rich 30 |
0.0002 | 0.0105 | 2.024 | PTPN20B | protein tyrosine phosphatase, non-receptor type 20B |
0.0002 | 0.0105 | 3.008 | SLC5A8 | solute carrier family 5 (sodium/monocarboxylate cotransporter), member 8 |
0.0002 | 0.0105 | 3.126 | SSMEM1 | serine-rich single-pass membrane protein 1 |
0.0002 | 0.0108 | 2.933 | CD28 | CD28 molecule |
0.0002 | 0.0108 | 3.91 | FLG2 | filaggrin family member 2 |
0.0002 | 0.0108 | 3.442 | IL2RA | interleukin 2 receptor, alpha |
0.0002 | 0.0108 | 1.794 | LDHB | lactate dehydrogenase B |
0.0002 | 0.0108 | 4.586 | OR10G2 | olfactory receptor, family 10, subfamily G, member 2 |
0.0003 | 0.0111 | 1.206 | AQP4 | aquaporin 4 |
0.0003 | 0.0111 | 1.499 | CAMK1D | calcium/calmodulin-dependent protein kinase ID |
0.0003 | 0.0111 | 1.851 | CSRNP3 | cysteine-serine-rich nuclear protein 3 |
0.0002 | 0.0111 | 4.068 | DPF3 | D4, zinc and double PHD fingers, family 3 |
0.0003 | 0.0111 | 1.121 | EIF2AK2 | eukaryotic translation initiation factor 2-alpha kinase 2 |
0.0002 | 0.0111 | 1.716 | HEATR9 | HEAT repeat containing 9 |
0.0003 | 0.0111 | 5.028 | LAMP3 | lysosomal-associated membrane protein 3 |
0.0003 | 0.0111 | 1.241 | MAPK13 | mitogen-activated protein kinase 13 |
0.0003 | 0.0111 | 2.144 | NAA11 | N(alpha)-acetyltransferase 11, NatA catalytic subunit |
0.0003 | 0.0111 | 2.652 | OR4D5 | olfactory receptor, family 4, subfamily D, member 5 |
0.0003 | 0.0111 | 2.904 | OR4M1 | olfactory receptor, family 4, subfamily M, member 1 |
0.0003 | 0.0111 | 4.603 | SIGLEC12 | sialic acid binding Ig-like lectin 12 (gene/pseudogene) |
0.0003 | 0.0111 | 1.359 | SLC1A2 | solute carrier family 1 (glial high affinity glutamate transporter), member 2 |
0.0003 | 0.0111 | 2.832 | TMA7 | translation machinery associated 7 homolog (S. cerevisiae) |
0.0003 | 0.0117 | 2.123 | TMEM211 | transmembrane protein 211 |
0.0003 | 0.0120 | 1.175 | B4GALT1 | UDP-Gal:betaGlcNAc beta 1,4- galactosyltransferase, polypeptide 1 |
0.0003 | 0.0120 | 1.968 | CYP3A5 | cytochrome P450, family 3, subfamily A, polypeptide 5 |
0.0003 | 0.0120 | 1.982 | GLRA1 | glycine receptor, alpha 1 |
0.0003 | 0.0120 | −1.739 | HBQ1 | hemoglobin, theta 1 |
0.0003 | 0.0120 | 1.956 | NDUFV2 | NADH dehydrogenase (ubiquinone) flavoprotein 2, 24kDa |
0.0003 | 0.0120 | 1.276 | PLCXD2 | phosphatidylinositol-specific phospholipase C, X domain containing 2 |
0.0003 | 0.0120 | 1.124 | RNF152 | ring finger protein 152 |
0.0003 | 0.0120 | 2.255 | SLC19A3 | solute carrier family 19 (thiamine transporter), member 3 |
0.0003 | 0.0120 | 1.334 | SLC4A4 | solute carrier family 4 (sodium bicarbonate cotransporter), member 4 |
0.0003 | 0.0121 | 1.201 | OR1F1 | olfactory receptor, family 1, subfamily F, member 1 |
0.0004 | 0.0123 | 2.899 | CXCL10 | chemokine (C-X-C motif) ligand 10 |
0.0004 | 0.0124 | 4.338 | CLDN8 | claudin 8 |
0.0004 | 0.0124 | 1.494 | SLC24A2 | solute carrier family 24 (sodium/potassium/calcium exchanger), member 2 |
0.0004 | 0.0126 | 1.841 | RPS2 | ribosomal protein S2 |
0.0004 | 0.0127 | 3.004 | CCL23 | chemokine (C-C motif) ligand 23 |
0.0004 | 0.0127 | 3.349 | IL7 | interleukin 7 |
0.0004 | 0.0128 | 1.574 | AANAT | aralkylamine N-acetyltransferase |
0.0004 | 0.0128 | 1.607 | DMD | dystrophin |
0.0004 | 0.0128 | 1.546 | GRIN2B | glutamate receptor, ionotropic, N-methyl D-aspartate 2B |
0.0004 | 0.0128 | 2.285 | MRPL18 | mitochondrial ribosomal protein L18 |
0.0004 | 0.0128 | 3.19 | OR10Q1 | olfactory receptor, family 10, subfamily Q, member 1 |
0.0004 | 0.0128 | 1.127 | TMEM220 | transmembrane protein 220 |
0.0004 | 0.0131 | 2.909 | CCR4 | chemokine (C-C motif) receptor 4 |
0.0005 | 0.0132 | 1.3 | ADD3 | adducin 3 (gamma) |
0.0005 | 0.0132 | 2.062 | ATP5J2 | ATP synthase, H+ transporting, mitochondrial Fo complex, subunit F2 |
0.0005 | 0.0132 | 3.716 | CCL20 | chemokine (C-C motif) ligand 20 |
0.0005 | 0.0132 | 2.559 | CD84 | CD84 molecule |
0.0004 | 0.0132 | 1.436 | CLCN6 | chloride channel, voltage-sensitive 6 |
0.0004 | 0.0132 | 1.107 | GIMAP2 | GTPase, IMAP family member 2 |
0.0005 | 0.0132 | 1.28 | KDM5A | lysine (K)-specific demethylase 5A |
0.0004 | 0.0132 | 1.216 | PPP1R16B | protein phosphatase 1, regulatory subunit 16B |
0.0005 | 0.0132 | 1.788 | RPL23A | ribosomal protein L23a |
0.0005 | 0.0132 | 1.329 | RPL41 | ribosomal protein L41 |
0.0005 | 0.0132 | 1.295 | SESN3 | sestrin 3 |
0.0004 | 0.0132 | 3.293 | SLC26A3 | solute carrier family 26 (anion exchanger), member 3 |
0.0005 | 0.0132 | 1.391 | SLC9A7 | solute carrier family 9, subfamily A (NHE7, cation proton antiporter 7), member 7 |
0.0005 | 0.0132 | 2.003 | SLC9C1 | solute carrier family 9, subfamily C (Na+-transporting carboxylic acid decarboxylase), member 1 |
0.0005 | 0.0132 | 1.475 | TAZ | tafazzin |
0.0005 | 0.0132 | 2.221 | TTR | transthyretin |
0.0005 | 0.0132 | 2.383 | WEE2 | WEE1 homolog 2 (S. pombe) |
0.0005 | 0.0133 | 3.245 | TSPO2 | translocator protein 2 |
0.0005 | 0.0134 | 4.467 | OR5L1 | olfactory receptor, family 5, subfamily L, member 1 |
0.0005 | 0.0134 | 2.422 | SLC6A2 | solute carrier family 6 (neurotransmitter transporter), member 2 |
0.0005 | 0.0139 | 1.609 | KLHL11 | kelch-like family member 11 |
0.0006 | 0.0140 | 1.336 | OR6X1 | olfactory receptor, family 6, subfamily X, member 1 |
0.0006 | 0.0142 | −1.685 | HBA1/HBA2 | hemoglobin, alpha 1 |
0.0006 | 0.0142 | 1.577 | INSM2 | insulinoma-associated 2 |
0.0006 | 0.0142 | 1.416 | RPL24 | ribosomal protein L24 |
0.0006 | 0.0149 | 1.341 | AR | androgen receptor |
0.0007 | 0.0157 | 1.547 | KSR2 | kinase suppressor of ras 2 |
0.0007 | 0.0159 | 1.474 | LY6G6C | lymphocyte antigen 6 complex, locus G6C |
0.0007 | 0.0159 | 1.813 | SAMD8 | sterile alpha motif domain containing 8 |
0.0007 | 0.0162 | 1.338 | CBL | Cbl proto-oncogene, E3 ubiquitin protein ligase |
0.0007 | 0.0162 | 2.951 | MEOX2 | mesenchyme homeobox 2 |
0.0007 | 0.0162 | 1.132 | PDE10A | phosphodiesterase 10A |
0.0007 | 0.0163 | 2.957 | COX6B2 | cytochrome c oxidase subunit VIb polypeptide 2 (testis) |
0.0007 | 0.0163 | 1.786 | GPR128 | G protein-coupled receptor 128 |
0.0007 | 0.0165 | 1.111 | ITGAV | integrin, alpha V |
0.0007 | 0.0165 | 1.719 | ZNF460 | zinc finger protein 460 |
0.0008 | 0.0167 | 1.198 | TNFRSF8 | tumor necrosis factor receptor superfamily, member 8 |
0.0008 | 0.0169 | 1.103 | TTLL7 | tubulin tyrosine ligase-like family, member 7 |
0.0008 | 0.0172 | 1.607 | CDKL5 | cyclin-dependent kinase-like 5 |
0.0008 | 0.0172 | 2.582 | CEACAM5 | carcinoembryonic antigen-related cell adhesion molecule 5 |
0.0008 | 0.0172 | 1.339 | MGAT4A | mannosyl (alpha-1,3-)-glycoprotein beta-1,4-N-acetylglucosaminyltransferase, isozyme A |
0.0008 | 0.0172 | 1.953 | NT5DC3 | 5′-nucleotidase domain containing 3 |
0.0008 | 0.0172 | 1.798 | PTGER2 | prostaglandin E receptor 2 (subtype EP2), 53kDa |
0.0008 | 0.0173 | 1.184 | CCR1 | chemokine (C-C motif) receptor 1 |
0.0008 | 0.0177 | 3.888 | PAPOLB | poly(A) polymerase beta (testis specific) |
0.0009 | 0.0178 | 1.109 | ADAT2 | adenosine deaminase, tRNA-specific 2 |
0.0009 | 0.0178 | 4.485 | HOXC4 | homeobox C4 |
0.0009 | 0.0178 | 2.925 | LPHN3 | latrophilin 3 |
0.0009 | 0.0178 | −2.549 | SPAG17 | sperm associated antigen 17 |
0.0009 | 0.0178 | 1.237 | THSD7A | thrombospondin, type I, domain containing 7A |
0.0009 | 0.0179 | 1.365 | DDX6 | DEAD (Asp-Glu-Ala-Asp) box helicase 6 |
0.0009 | 0.0179 | 1.164 | PIWIL2 | piwi-like RNA-mediated gene silencing 2 |
0.0009 | 0.0180 | 1.232 | DENND5B | DENN/MADD domain containing 5B |
0.0009 | 0.0180 | 4.287 | TAAR6 | trace amine associated receptor 6 |
0.0009 | 0.0181 | 1.217 | KIAA1324 | KIAA1324 |
0.0009 | 0.0181 | 1.683 | RYR1 | ryanodine receptor 1 (skeletal) |
0.0009 | 0.0182 | 1.419 | TLR6 | toll-like receptor 6 |
0.0010 | 0.0183 | 1.99 | ZNF593 | zinc finger protein 593 |
0.0010 | 0.0184 | 1.454 | MIB1 | mindbomb E3 ubiquitin protein ligase 1 |
0.0010 | 0.0185 | 1.12 | SLC35F1 | solute carrier family 35, member F1 |
0.0010 | 0.0187 | 1.885 | APLNR | apelin receptor |
0.0010 | 0.0187 | 1.319 | CAV1 | caveolin 1, caveolae protein, 22kDa |
0.0010 | 0.0187 | 1.204 | IL1B | interleukin 1, beta |
0.0010 | 0.0187 | 1.228 | LONRF2 | LON peptidase N-terminal domain and ring finger 2 |
0.0010 | 0.0187 | 1.261 | TAOK1 | TAO kinase 1 |
0.0011 | 0.0189 | 1.247 | EXOC6B | exocyst complex component 6B |
0.0011 | 0.0192 | −1.265 | SNRPD2 | small nuclear ribonucleoprotein D2 polypeptide 16.5kDa |
0.0011 | 0.0193 | 2.165 | MT1L | metallothionein 1L (gene/pseudogene) |
0.0011 | 0.0193 | 1.142 | NFAT5 | nuclear factor of activated T-cells 5, tonicity-responsive |
0.0011 | 0.0193 | 1.145 | NRIP1 | nuclear receptor interacting protein 1 |
0.0011 | 0.0194 | 1.954 | FABP12 | fatty acid binding protein 12 |
0.0011 | 0.0194 | 1.104 | GRINA | glutamate receptor, ionotropic, N-methyl D-aspartate-associated protein 1 (glutamate binding) |
0.0011 | 0.0194 | 1.263 | HIPK3 | homeodomain interacting protein kinase 3 |
0.0012 | 0.0202 | 2.898 | SCGB1D2 | secretoglobin, family 1D, member 2 |
0.0012 | 0.0206 | 1.984 | TNFRSF9 | tumor necrosis factor receptor superfamily, member 9 |
0.0013 | 0.0211 | 1.523 | RAB11FIP4 | RAB11 family interacting protein 4 (class II) |
0.0014 | 0.0220 | −1.495 | ACTB | actin, beta |
0.0014 | 0.0220 | 1.13 | BTBD9 | BTB (POZ) domain containing 9 |
0.0014 | 0.0220 | 1.491 | PANK3 | pantothenate kinase 3 |
0.0014 | 0.0220 | 1.991 | SERF2 | small EDRK-rich factor 2 |
0.0014 | 0.0222 | −2.489 | CGB | chorionic gonadotropin, beta polypeptide |
0.0014 | 0.0222 | 1.908 | LTB4R2 | leukotriene B4 receptor 2 |
0.0015 | 0.0223 | 1.618 | ACR | acrosin |
0.0015 | 0.0223 | 1.109 | APPBP2 | amyloid beta precursor protein (cytoplasmic tail) binding protein 2 |
0.0015 | 0.0223 | 1.83 | CXCR2 | chemokine (C-X-C motif) receptor 2 |
0.0015 | 0.0223 | 1.153 | GMFB | glia maturation factor, beta |
0.0015 | 0.0223 | 1.113 | ZBED6 | zinc finger, BED-type containing 6 |
0.0016 | 0.0224 | 1.703 | B3GALT5 | UDP-Gal:betaGlcNAc beta 1,3-galactosyltransferase, polypeptide 5 |
0.0015 | 0.0224 | 1.484 | DBNL | drebrin-like |
0.0016 | 0.0224 | 2.839 | OR7D4 | olfactory receptor, family 7, subfamily D, member 4 |
0.0016 | 0.0224 | 2.077 | SUMO2 | small ubiquitin-like modifier 2 |
0.0016 | 0.0226 | 1.985 | PIWIL3 | piwi-like RNA-mediated gene silencing 3 |
0.0016 | 0.0226 | 1.108 | SLC38A2 | solute carrier family 38, member 2 |
0.0016 | 0.0227 | 1.505 | DTX3L | deltex 3 like, E3 ubiquitin ligase |
0.0016 | 0.0227 | 2.99 | MS4A15 | membrane-spanning 4-domains, subfamily A, member 15 |
0.0017 | 0.0229 | 1.268 | DRAM1 | DNA-damage regulated autophagy modulator 1 |
0.0017 | 0.0232 | 1.234 | CNOT6 | CCR4-NOT transcription complex, subunit 6 |
0.0017 | 0.0232 | 1.606 | SERPINA10 | serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 10 |
0.0017 | 0.0235 | 1.652 | FAT1 | FAT atypical cadherin 1 |
0.0018 | 0.0239 | 1.606 | FAM26F | family with sequence similarity 26, member F |
0.0018 | 0.0239 | 1.717 | LCN15 | lipocalin 15 |
0.0018 | 0.0240 | 1.233 | PRR14L | proline rich 14-like |
0.0018 | 0.0242 | 1.553 | TRPC4 | transient receptor potential cation channel, subfamily C, member 4 |
0.0019 | 0.0249 | 2.268 | MUC6 | mucin 6, oligomeric mucus/gel-forming |
0.0020 | 0.0251 | 1.177 | LIN7C | lin-7 homolog C (C. elegans) |
0.0020 | 0.0251 | 1.207 | MGAT4B | mannosyl (alpha-1,3-)-glycoprotein beta-1,4-N-acetylglucosaminyltransferase, isozyme B |
0.0020 | 0.0252 | 1.489 | HTR1F | 5-hydroxytryptamine (serotonin) receptor 1F, G protein-coupled |
0.0021 | 0.0256 | 2.431 | SDHC | succinate dehydrogenase complex, subunit C, integral membrane protein, 15kDa |
0.0021 | 0.0256 | 1.136 | SNX27 | sorting nexin family member 27 |
0.0023 | 0.0267 | 1.119 | DPP8 | dipeptidyl-peptidase 8 |
0.0022 | 0.0267 | 1.613 | DUSP19 | dual specificity phosphatase 19 |
0.0022 | 0.0267 | 1.953 | LRFN3 | leucine rich repeat and fibronectin type III domain containing 3 |
0.0023 | 0.0267 | 1.166 | PLXNA4 | plexin A4 |
0.0022 | 0.0267 | 1.12 | PRSS44 | protease, serine, 44 |
0.0022 | 0.0267 | 1.967 | TAS2R1 | taste receptor, type 2, member 1 |
0.0023 | 0.0268 | 1.998 | XIAP | X-linked inhibitor of apoptosis |
0.0023 | 0.0269 | 1.317 | C21orf91 | chromosome 21 open reading frame 91 |
0.0023 | 0.0269 | 1.223 | TMEM170B | transmembrane protein 170B |
0.0024 | 0.0270 | 1.462 | GUCY1A2 | guanylate cyclase 1, soluble, alpha 2 |
0.0024 | 0.0270 | 1.275 | SLC5A3 | solute carrier family 5 (sodium/myo-inositol cotransporter), member 3 |
0.0024 | 0.0274 | 1.702 | KIAA2018 | KIAA2018 |
0.0024 | 0.0275 | 1.121 | PDCD1LG2 | programmed cell death 1 ligand 2 |
0.0025 | 0.0278 | 1.378 | MPV17L | MPV17 mitochondrial membrane protein-like |
0.0026 | 0.0283 | 1.126 | LRRC8C | leucine rich repeat containing 8 family, member C |
0.0028 | 0.0291 | 1.117 | C1QTNF9 | C1q and tumor necrosis factor related protein 9 |
0.0028 | 0.0291 | 1.102 | SMOC2 | SPARC related modular calcium binding 2 |
0.0029 | 0.0296 | 1.513 | MAGT1 | magnesium transporter 1 |
0.0029 | 0.0296 | −1.158 | OBP2A | odorant binding protein 2A |
0.0029 | 0.0297 | 1.472 | NRCAM | neuronal cell adhesion molecule |
0.0029 | 0.0298 | 1.172 | RPL35A | ribosomal protein L35a |
0.0030 | 0.0299 | 1.552 | DOK2 | docking protein 2, 56kDa |
0.0031 | 0.0303 | 1.271 | ADH1A | alcohol dehydrogenase 1A (class I), alpha polypeptide |
0.0032 | 0.0303 | −1.435 | NPIPA1 | nuclear pore complex interacting protein family, member A1 |
0.0033 | 0.0307 | 1.688 | PPARA | peroxisome proliferator-activated receptor alpha |
0.0034 | 0.0312 | 1.197 | CX3CR1 | chemokine (C-X3-C motif) receptor 1 |
0.0034 | 0.0312 | 1.106 | RALGPS2 | Ral GEF with PH domain and SH3 binding motif 2 |
0.0035 | 0.0312 | 1.749 | TMPPE | transmembrane protein with metallophosphoesterase domain |
0.0035 | 0.0313 | 2.607 | SIRPD | signal-regulatory protein delta |
0.0035 | 0.0314 | 1.458 | LCLAT1 | lysocardiolipin acyltransferase 1 |
0.0035 | 0.0314 | 1.39 | LMF1 | lipase maturation factor 1 |
0.0035 | 0.0314 | 1.475 | TCP11 | t-complex 11, testis-specific |
0.0037 | 0.0319 | 1.251 | CYP7B1 | cytochrome P450, family 7, subfamily B, polypeptide 1 |
0.0037 | 0.0319 | 1.301 | RFX3 | regulatory factor X, 3 (influences HLA class II expression) |
0.0037 | 0.0319 | 1.448 | THSD7B | thrombospondin, type I, domain containing 7B |
0.0036 | 0.0319 | 2.124 | VBP1 | von Hippel-Lindau binding protein 1 |
0.0040 | 0.0332 | 1.258 | CCDC79 | coiled-coil domain containing 79 |
0.0040 | 0.0332 | 1.72 | CD5L | CD5 molecule-like |
0.0040 | 0.0332 | 1.841 | SLC26A2 | solute carrier family 26 (anion exchanger), member 2 |
0.0039 | 0.0332 | 1.17 | TLR3 | toll-like receptor 3 |
0.0040 | 0.0335 | 1.151 | PTGFR | prostaglandin F receptor (FP) |
0.0042 | 0.0339 | 2.154 | SNTB2 | syntrophin, beta 2 (dystrophin-associated protein A1, 59kDa, basic component 2) |
0.0043 | 0.0343 | 1.237 | KAT7 | K(lysine) acetyltransferase 7 |
0.0043 | 0.0343 | 1.583 | POU1F1 | POU class 1 homeobox 1 |
0.0045 | 0.0355 | −2.002 | LRRC71 | leucine rich repeat containing 71 |
0.0046 | 0.0356 | 1.155 | OVGP1 | oviductal glycoprotein 1, 120kDa |
0.0046 | 0.0357 | −1.212 | C1orf158 | chromosome 1 open reading frame 158 |
0.0046 | 0.0357 | 1.311 | ENPP6 | ectonucleotide pyrophosphatase/phosphodiesterase 6 |
0.0046 | 0.0357 | 1.251 | TIPARP | TCDD-inducible poly(ADP-ribose) polymerase |
0.0047 | 0.0358 | 1.334 | ZNF280B | zinc finger protein 280B |
0.0047 | 0.0359 | 1.255 | UNC5C | unc-5 homolog C (C. elegans) |
0.0048 | 0.0364 | 1.381 | FNIP1 | folliculin interacting protein 1 |
0.0049 | 0.0368 | −1.177 | RPL19 | ribosomal protein L19 |
0.0049 | 0.0368 | 1.116 | SLC5A7 | solute carrier family 5 (sodium/choline cotransporter), member 7 |
0.0050 | 0.0369 | 1.121 | MDM4 | MDM4, p53 regulator |
0.0050 | 0.0371 | 1.592 | EXPH5 | exophilin 5 |
0.0050 | 0.0373 | 1.73 | ALG10 | ALG10, alpha-1,2-glucosyltransferase |
0.0052 | 0.0380 | 1.521 | SNX30 | sorting nexin family member 30 |
0.0054 | 0.0386 | 1.639 | ILDR1 | immunoglobulin-like domain containing receptor 1 |
0.0054 | 0.0386 | 1.253 | RPL34 | ribosomal protein L34 |
0.0054 | 0.0387 | −1.959 | GALNT8 | polypeptide N-acetylgalactosaminyltransferase 8 |
0.0056 | 0.0390 | 1.231 | CMTM4 | CKLF-like MARVEL transmembrane domain containing 4 |
0.0056 | 0.0390 | 1.839 | HIST1H4E | histone cluster 1, H4e |
0.0056 | 0.0390 | 1.951 | NSL1 | NSL1, MIS12 kinetochore complex component |
0.0057 | 0.0393 | 1.251 | RBM12B-AS1 | RBM12B antisense RNA 1 |
0.0058 | 0.0396 | 1.449 | CACNA1E | calcium channel, voltage-dependent, R type, alpha 1E subunit |
0.0059 | 0.0399 | 1.377 | ITGA1 | integrin, alpha 1 |
0.0060 | 0.0399 | 1.221 | ZFP14 | ZFP14 zinc finger protein |
0.0061 | 0.0403 | 1.343 | RAB27B | RAB27B, member RAS oncogene family |
0.0065 | 0.0417 | 2.172 | ATXN1 | ataxin 1 |
0.0067 | 0.0425 | 1.141 | NUP155 | nucleoporin 155kDa |
0.0067 | 0.0425 | 1.11 | SERINC5 | serine incorporator 5 |
0.0067 | 0.0425 | 1.74 | ZNF529 | zinc finger protein 529 |
0.0070 | 0.0436 | 1.66 | CCDC68 | coiled-coil domain containing 68 |
0.0074 | 0.0449 | 1.507 | MTTP | microsomal triglyceride transfer protein |
0.0078 | 0.0459 | 1.81 | KRT23 | keratin 23 (histone deacetylase inducible) |
0.0077 | 0.0459 | 1.222 | ZDHHC21 | zinc finger, DHHC-type containing 21 |
0.0080 | 0.0465 | 1.452 | COMMD9 | COMM domain containing 9 |
0.0088 | 0.0491 | 1.112 | ANKRD20A4 | ankyrin repeat domain 20 family, member A4 |
0.0089 | 0.0496 | 1.201 | TTC39B | tetratricopeptide repeat domain 39B |
Supplementary material can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:...
Figure 2.
Volcano plots comparing the normalized expression of genes in long-term cocaine self-administering animals to yoked saline controls in the ventral tegmental area (A) and nucleus accumbens (B).
Using these lists of dysregulated genes, further analysis focusing on the identification of common or shared pathways was undertaken using Ingenuity Pathway Analysis. In the VTA, the most notable differences were found in pathways associated with dopaminergic signaling (Figure 3A). This pathway, as well as others that highly overlap (including catecholamine biosynthesis and serotonin signaling), is significantly enriched for differentially expressed genes. In the NAc, many of the enriched pathways were associated with immune response, broadly or specifically, and cellular responses to stress (Figure 3B). These findings were broad-based and robust to methodologies for assigning differential expression.
Figure 3.
Canonical pathway enrichment analysis of genes differentially expressed in the VTA (A) and NAc (B) of cocaine-self-administering animals. Significance values (dashed lines; p < 0.05, -log(p-value) > 1.3) are determined through IPA default methodologies. The ratio of genes differentially expressed to the total number found in the pathway is also shown.
3.3 Expression differences in the VTA
The most notable differences in the VTA (Figure 4A) of animals exposed to cocaine were found in the dopaminergic pathway. Both tyrosine hydroxylase (TH), responsible for converting L-tyrosine to L-3,4-dihydroxyphenylalanine (L-DOPA), and dopa decarboxylase (DDC), responsible for the conversion of L-DOPA to dopamine, were down-regulated (q < 0.05). And while there is also a robust down-regulation of the dopamine transporter (SLC6A3; q < 0.01), there were no significant differences in expression of any of the dopamine receptors. These findings confirm previous studies in post-mortem human cocaine abusers (Bannon et al., 2014; Bannon et al., 2015; Little et al., 1998; Zhou et al., 2014a) as well as in rats following extended cocaine exposure (Cerruti et al., 1994; Letchworth et al., 1997; Letchworth et al., 1999).
Figure 4.
Heatmap showing the effects of long-term cocaine self-administration on gene expression in the ventral tegmental area (A) and nucleus accumbens (B). Higher expression levels are indicated in red and lower expression levels in yellow. Cocaine and saline animals are grouped together.
In rhesus macaques exposed to cocaine, the transcription factors FOXA1 and FOXA2 were both down regulated (q < 0.05 and q < 0.01 respectively). These genes have previously been shown to specify midbrain dopaminergic fate (Ferri et al., 2007) and are direct regulators not only of DDC and TH in mature dopaminergic neurons, but also of engrailed 1 and 2 (EN1, EN2) and the nuclear receptor NR4A2 (also known as NURR1) in immature neurons. FOXA2 down regulation has likewise been observed in the midbrain of human cocaine abusers (Bannon et al., 2014; Bannon et al., 2015). In addition to developmental roles in specifying dopaminergic neuronal fate, FOXA1 and FOXA2 have also been demonstrated to regulate midbrain dopaminergic function and survival in the adult brain (Kittappa et al., 2007).
3.4 Expression differences in the NAc
Dominating the differences observed in the NAc (Figure 4B) were genes associated with immune response and inflammation. This included numerous cytokines (IL1B, CXCL1, CXCL8 (IL8), CXCL10, CCL20, CCL23) and receptors (CXCR2, CCR1). We also see significant differences in expression of several toll-like receptors (TLR3, TLR6, TLR10). These pathways have previously been associated with a neuroprotective response to stress modulated as well by EIF2AK2, also known as the dsRNA responsive protein kinase PKR (Hsu et al., 2004). This gene was also differentially expressed in the NAc of cocaine administering animals (q < 0.05). This finding extends to genes downstream of PKR that also show significant differences in cocaine-exposed animals (EIF2 signaling; Figure 3B). These results show strong evidence reflective of an inflammatory response coinciding with cellular stress and neuronal death.
A large number of other genes of diverse function were also upregulated. This included a large number of genes that were not detected in the saline-yoked animals, but that were detected at extremely low, but non-zero, levels in the cocaine animals. This, perhaps, can be explained in part through the effects of chromatin remodeling. H3 and H4 were both differentially expressed (q < 0.005 and q < 0.05 respectively). It is hypothesized that a large amount of the upregulated genes observed here, particularly those that have near absent expression levels in saline control animals, are the result of “leaky” expression resulting from epigenetic changes and resulting chromatin remodeling. It is notable that at least one gene, KRT23, unrelated to brain function that was upregulated (q < 0.05) has previously been demonstrated to be highly inducible through histone hyperacetylation (Zhang et al., 2001). While this does not preclude a functional relevance for many of these specific expression changes, it may represent a symptom rather than an underlying causal factor.
4. Discussion
Under conditions of limited access (1 hr/day) and long-term exposure (approximately 100 consecutive days), we observed a general increase over time in cocaine self-administration in monkeys. However, while statistically significant, this escalation was modest in size. While there was a limit on the total amount of cocaine that was available for the animals to consume (so as to not lead to overdose), this did not fully account for the magnitude effect. Rather, what was observed was a relatively robust increase in the rate at which cocaine was consumed early in the daily sessions. In later sessions not only do animals consume, on average, more cocaine (roughly 30% increase over the 100 days), but their consumption increases more than 80% in the first 15 minutes while decreasing 15% in the remaining 45 minutes.
The increase in cocaine taking across sessions has some similarities to the ”escalation” phenomenon documented in rats self-administering cocaine for prolonged periods (Edwards and Koob, 2013). Specifically, animals increased their consumption over time and this consumption began to occur more heavily earlier in the session. Although our findings may reflect a similar escalation effect, there were notable differences. First, the increase in self-administration was observed with relatively short (1 hr) session lengths and manifested primarily as an increase in the number of sessions in which the maximum amount of cocaine was taken (escalation studies in rats typically do not limit the number of infusions available in a session). Second, the increase in cocaine taking did not “ramp up” over the first week of self-administration as seen with rats (Edwards and Koob, 2013), rather the increase was much more gradual over time. Regardless, an increase in the amount of cocaine consumed with increasing experience is a hallmark of human cocaine addiction that can be modeled in nonhuman subjects and it may be the case that the differences in magnitude and pattern of escalation across species merely result from as-yet fully defined parametric variables.
It is also important to note that prior studies on escalation of cocaine taking using rhesus macaques as subjects generally did not observe consistent increases in self-administration over time with either marked individual differences in the extent to which escalation occurred or no change in self-administration across sessions (Czoty et al., 2007; Henry and Howell, 2009; Kirkland Henry et al., 2009). There are many differences among the studies (e.g., schedule of reinforcement, number of sessions) and it is unclear at present which factor is the primary determinant of the extent to which escalation occurs in nonhuman primates. Nevertheless, modelling this important feature of human addictive behavior suggests an important avenue into translationally relevant interventions.
Developing an animal model that faithfully recapitulates the human disease state is important because of our increased levels of experimental control. In addition to a much greater understanding of the exact nature of exposure to drug, we have a much more precise and meaningful opportunity to look at specifically associated molecular effects. The neuroanatomy, genetics, and physiology of the brain of a rhesus macaque are highly similar to a human, but the specifics of drug exposure are better defined and tissue collection post-mortem is much more rapid. This allows us to compare the findings here in the animal model to post-mortem human studies and be confident of the direct causal relationships between drug use and brain molecular differences.
The initial focus of these studies was on the mesolimbic system, specifically the ventral tegmental area and the nucleus accumbens. This has been the traditional focus for addiction studies and it allows for direct comparisons with previous literature. Differences observed in gene expression in the ventral tegmental area have generally centered on the dopaminergic system. Indeed, in this work we also see a down-regulation of genes associated with dopaminergic neuron development and function, including the genes traditionally associated with dopamine synthesis (DDH and TH) and transport (SLC6A3), consistent with findings from human cocaine abusers. Previous studies have observed decreases in the dopamine transporter in post-mortem studies from human cocaine abusers (Bannon et al., 2014; Bannon et al., 2015; Little et al., 1998; Zhou et al., 2014a) as well as in rats following extended cocaine exposure (Cerruti et al., 1994; Letchworth et al., 1997; Letchworth et al., 1999; Zhang et al., 2012). The parallels between these studies and previous human studies extend to a reduced expression of tyrosine hydroxylase in the VTA (Bannon et al., 2014; Bannon et al., 2015), although rat studies have been more ambiguous (Freeman et al., 2000; Rodriguez-Espinosa and Fernandez-Espejo, 2015; Vrana et al., 1993).
We also see corresponding differences in the upstream regulators of these genes, notably FOXA2, that are similar to human post-mortem findings (Bannon et al., 2014; Bannon et al., 2015). FOXA1 and FOXA2 have previously been demonstrated to regulate midbrain dopaminergic development and function (Ferri et al., 2007; Kittappa et al., 2007; Lin et al., 2009; Metzakopian et al., 2015; Metzakopian et al., 2012; Stott et al., 2013). While there does seems to be a developmental role for FOXA1 and FOXA2 (Hallonet et al., 2002; Mavromatakis et al., 2011; Nakatani et al., 2010), it is their role in maintaining dopaminergic identity in mature neurons that is likely more relevant to studies of cocaine effects. Perhaps most notable beyond this general down-regulation of dopaminergic tone, is the absence of evidence of any other major coordinated effects. While it certainly remains that individual differences in gene expression detected may be important, the primary finding of this study emphasizes the central role of changes in the dopamine system in the VTA. While other effects cannot be ruled out, this and other studies place dopaminergic dysregulation centrally.
The NAc in the present study is characterized mainly by differences associated with an activation of microglia akin to that seen in brain injury and chronic disease. This activation has been suggested to be important in the overall neurobiology of addiction (Crews et al., 2011). TLR-mediated pathways have previously been demonstrated to have proinflammatory neuroprotective effects on astrocytes (Bsibsi et al., 2006). Moreover, this immune response also has been previously demonstrated to be involved in the remyelination process (Glezer et al., 2006). Consistent with these observations, a number of post-mortem human studies have reported myelin dysregulation in the NAc (Albertson et al., 2004; Bannon et al., 2005), although we see little evidence for demyelination here, possibly as a result of the timing of drug exposure. Although this work does not address whether the activation of innate immunity is solely a response to insult or if these changes are driving the addiction process, it does demonstrate an important, region specific, role for immune signaling that further builds upon an existing neuroimmune hypothesis of addiction.
Coupled with the immune activation, was a widespread up-regulation of a diverse array of genes in the NAc, including a significant subset that show no expression in unexposed animals and are not generally expressed in brain tissue. This is hypothesized to be attributable to cocaine-mediated chromatin remodeling, accounting for both the diversity of genes as well as the overrepresentation of upregulation compared to downregulation. A substantial literature exists identifying epigenomic changes in the NAc following long-term cocaine exposure specifically and addiction generally (Nestler, 2014; Sadri-Vakili, 2014). These epigenetic changes have been shown to correlate with drug-induced behavioral change and neuronal plasticity (Schmidt et al., 2013). In essence, chromatin is thought to be moving from a “closed” state in which transcription is tightly repressed, to an “open” state where more transcriptional activity is occurring. Cocaine mediated chromatin remodeling in the NAc has been previously demonstrated (Feng et al., 2014; Kumar et al., 2005; Maze et al., 2011) and the role of chromatin remodeling in gene expression is under increasing scrutiny (Bell et al., 2011; Mirabella et al., 2016). In particular, the role of chromatin reorganization in macrophage response is increasingly being recognized with open chromatin and high gene expression consistent with activation (Glass and Natoli, 2016; Schmidt et al., 2016). The indirect evidence here for epigenetic change not only includes this broad upregulation of gene expression but also differences in expression of multiple genes encoding histones.
Epigenetic changes have previously been suggested to correlate with the transition from use to abuse (Schmidt et al., 2013). It is perhaps notable, then, that the differences we observe here seem to be much more pronounced in the NAc and less so in the VTA, further bolstering the concept that while the VTA is associated with the initial and acute action of the drug, it is the NAc that is associated with longer-term neuro-adaptations (Koya et al., 2009). The picture emerging here is consistent with hypotheses that suggest dopaminergic changes in presynaptic neurons from the VTA lead to, or act in concert with, epigenetic remodeling and immune activation post-synaptically in the NAc. This model supports a central mechanism for developing addiction, independent of the specific mechanisms of action of drugs of abuse.
It is important to note, however, that this study is not exhaustive. While environmental variables, notably including cocaine administration, are controlled in these studies, sample sizes are necessarily small; together, these factors limit the effect size that it is possible to detect. While some inter-individual variability does exist in this study it is relatively minor and it is difficult to draw meaningful conclusions. The findings presented here do not exhaustively represent all the differences that occur with cocaine exposure, but rather a subset, likely of the largest effect sizes. It is meaningful that many of the key findings here are supported by, and supportive of, previous studies, particularly humans both with regards to behaviors associated with cocaine self-administration as well as gene expression responses.
Another limitation of the present study is the level of structural and cellular detail interrogated. In the NAc, differences exist between the core and shell (Brauer et al., 2000; Heimer et al., 1997; Martin and Cork, 2014; Meredith et al., 1996) that extend to circuitry and behavior (Baliki et al., 2013; Meredith et al., 2008; Saddoris et al., 2013), but these regions have been conflated in an effort to prioritize consistency between animals. Nor, have the cell-type distributions of the regions been explored. From the data, microglial activation is inferred in the NAc, but further studies that dissociate neuronal and glial effects are needed for confirmation. It is perhaps important to note, however, that the conflation of cell types or regions have the effect of reducing signal, effectively making the present study conservative.
In conclusion, this work demonstrates the power of an unbiased approach to transcriptomic profiling in a nonhuman primate model of long-term cocaine self-administration. Not only is this a meaningful and translationally-relevant model of human addiction, but it also affords added control of environmental variables and the ability to parse cause and effect in relationships between drug exposure and differences in gene expression.
Supplementary Material
Highlights.
Patterns of cocaine self-administration in rhesus macaques model humans
Gene expression differences are observed in the mesolimbic system after exposure
Changes in the ventral tegmental area are associated with the dopaminergic system
Changes in the nucleus accumbens reflect neuroinflammation and chromatin remodeling
Acknowledgments
This work was supported by grants from the NIH: DA021420 (WDY), OD011103.
Footnotes
Supplementary material can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:...
Supplementary material can be found by accessing the online version of this paper at http://dx.doi.org and by entering doi:...
Contributors
WDY and JKR were responsible for study concept and design. NMS and JKR were responsible for collection of animal data. WDY, SVM, DBG were responsible for brain and tissue collection. DBG and EJV were responsible for data analysis. EJV and JKR drafted the manuscript. All authors critically reviewed the manuscript and approved the final version for publication.
Conflict of Interest
No conflict declared.
Role of funding source
Nothing declared.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Albertson DN, Pruetz B, Schmidt CJ, Kuhn DM, Kapatos G, Bannon MJ. Gene expression profile of the nucleus accumbens of human cocaine abusers: Evidence for dysregulation of myelin. J Neurochem. 2004;88:1211–1219. doi: 10.1046/j.1471-4159.2003.02247.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baliki MN, Mansour A, Baria AT, Huang L, Berger SE, Fields HL, Apkarian AV. Parceling human accumbens into putative core and shell dissociates encoding of values for reward and pain. J Neurosci. 2013;33:16383–16393. doi: 10.1523/JNEUROSCI.1731-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bannon M, Kapatos G, Albertson D. Gene expression profiling in the brains of human cocaine abusers. Addict Biol. 2005;10:119–126. doi: 10.1080/13556210412331308921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bannon MJ, Johnson MM, Michelhaugh SK, Hartley ZJ, Halter SD, David JA, Kapatos G, Schmidt CJ. A molecular profile of cocaine abuse includes the differential expression of genes that regulate transcription, chromatin, and dopamine cell phenotype. Neuropsychopharmacol. 2014;39:2191–2199. doi: 10.1038/npp.2014.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bannon MJ, Savonen CL, Hartley ZJ, Johnson MM, Schmidt CJ. Investigating the potential influence of cause of death and cocaine levels on the differential expression of genes associated with cocaine abuse. PloS one. 2015;10:e0117580. doi: 10.1371/journal.pone.0117580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnett G, Hawks R, Resnick R. Cocaine pharmacokinetics in humans. J Ethnopharmacol. 1981;3:353–366. doi: 10.1016/0378-8741(81)90063-5. [DOI] [PubMed] [Google Scholar]
- Bell O, Tiwari VK, Thoma NH, Schubeler D. Determinants and dynamics of genome accessibility. Nat Rev Genet. 2011;12:554–564. doi: 10.1038/nrg3017. [DOI] [PubMed] [Google Scholar]
- Brauer K, Hausser M, Hartig W, Arendt T. The core-shell dichotomy of nucleus accumbens in the rhesus monkey as revealed by double-immunofluorescence and morphology of cholinergic interneurons. Brain Res. 2000;858:151–162. doi: 10.1016/s0006-8993(00)01938-7. [DOI] [PubMed] [Google Scholar]
- Bsibsi M, Persoon-Deen C, Verwer RW, Meeuwsen S, Ravid R, Van Noort JM. Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia. 2006;53:688–695. doi: 10.1002/glia.20328. [DOI] [PubMed] [Google Scholar]
- Bu Q, Hu Z, Chen F, Zhu R, Deng Y, Shao X, Li Y, Zhao J, Li H, Zhang B, Lv L, Yan G, Zhao Y, Cen X. Transcriptome analysis of long non-coding RNAs of the nucleus accumbens in cocaine-conditioned mice. J Neurochem. 2012;123:790–799. doi: 10.1111/jnc.12006. [DOI] [PubMed] [Google Scholar]
- Cerruti C, Pilotte NS, Uhl G, Kuhar MJ. Reduction in dopamine transporter mRNA after cessation of repeated cocaine administration. Brain Res Mol Brain Res. 1994;22:132–138. doi: 10.1016/0169-328x(94)90040-x. [DOI] [PubMed] [Google Scholar]
- Chen CL, Liu H, Guan X. Changes in microRNA expression profile in hippocampus during the acquisition and extinction of cocaine-induced conditioned place preference in rats. J Biomed Sci. 2013;20:96. doi: 10.1186/1423-0127-20-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crews FT, Zou J, Qin L. Induction of innate immune genes in brain create the neurobiology of addiction. Brain Behav Immun. 2011;25(Suppl 1):S4–S12. doi: 10.1016/j.bbi.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Czoty PW, Reboussin BA, Calhoun TL, Nader SH, Nader MA. Long-term cocaine self-administration under fixed-ratio and second-order schedules in monkeys. Psychopharmacol. 2007;191:287–295. doi: 10.1007/s00213-006-0665-z. [DOI] [PubMed] [Google Scholar]
- Edwards S, Koob GF. Escalation of drug self-administration as a hallmark of persistent addiction liability. Behav Pharmacol. 2013;24:356–362. doi: 10.1097/FBP.0b013e3283644d15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eipper-Mains JE, Kiraly DD, Duff MO, Horowitz MJ, McManus CJ, Eipper BA, Graveley BR, Mains RE. Effects of cocaine and withdrawal on the mouse nucleus accumbens transcriptome. Genes Brain Behav. 2013;12:21–33. doi: 10.1111/j.1601-183X.2012.00873.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng J, Wilkinson M, Liu X, Purushothaman I, Ferguson D, Vialou V, Maze I, Shao N, Kennedy P, Koo J, Dias C, Laitman B, Stockman V, LaPlant Q, Cahill ME, Nestler EJ, Shen L. Chronic cocaine-regulated epigenomic changes in mouse nucleus accumbens. Genome Biol. 2014;15:R65. doi: 10.1186/gb-2014-15-4-r65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferri AL, Lin W, Mavromatakis YE, Wang JC, Sasaki H, Whitsett JA, Ang SL. Foxa1 and Foxa2 regulate multiple phases of midbrain dopaminergic neuron development in a dosage-dependent manner. Development. 2007;134:2761–2769. doi: 10.1242/dev.000141. [DOI] [PubMed] [Google Scholar]
- Freeman WM, Yohrling GJ, Daunais JB, Gioia L, Hart SL, Porrino LJ, Davies HM, Vrana KE. A cocaine analog, 2beta-propanoyl-3beta-(4-tolyl)-tropane (PTT), reduces tyrosine hydroxylase in the mesolimbic dopamine pathway. Drug Alcohol Depend. 2000;61:15–21. doi: 10.1016/s0376-8716(00)00119-8. [DOI] [PubMed] [Google Scholar]
- Glass CK, Natoli G. Molecular control of activation and priming in macrophages. Nat Immunol. 2016;17:26–33. doi: 10.1038/ni.3306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glezer I, Lapointe A, Rivest S. Innate immunity triggers oligodendrocyte progenitor reactivity and confines damages to brain injuries. FASEB journal : Official publication of the FASEB J. 2006;20:750–752. doi: 10.1096/fj.05-5234fje. [DOI] [PubMed] [Google Scholar]
- Hallonet M, Kaestner KH, Martin-Parras L, Sasaki H, Betz UA, Ang SL. Maintenance of the specification of the anterior definitive endoderm and forebrain depends on the axial mesendoderm: A study using HNF3beta/Foxa2 conditional mutants. Dev Biol. 2002;243:20–33. doi: 10.1006/dbio.2001.0536. [DOI] [PubMed] [Google Scholar]
- Heimer L, Alheid GF, de Olmos JS, Groenewegen HJ, Haber SN, Harlan RE, Zahm DS. The accumbens: Beyond the core-shell dichotomy. J Neuropsychiatry Clin Neurosci. 1997;9:354–381. doi: 10.1176/jnp.9.3.354. [DOI] [PubMed] [Google Scholar]
- Henry PK, Howell LL. Cocaine-induced reinstatement during limited and extended drug access conditions in rhesus monkeys. Psychopharmacol. 2009;204:523–529. doi: 10.1007/s00213-009-1485-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hope B, Kosofsky B, Hyman SE, Nestler EJ. Regulation of immediate early gene expression and AP-1 binding in the rat nucleus accumbens by chronic cocaine. Proc Nat Acad Sci USA. 1992;89:5764–5768. doi: 10.1073/pnas.89.13.5764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu LC, Park JM, Zhang K, Luo JL, Maeda S, Kaufman RJ, Eckmann L, Guiney DG, Karin M. The protein kinase PKR is required for macrophage apoptosis after activation of Toll-like receptor 4. Nat. 2004;428:341–345. doi: 10.1038/nature02405. [DOI] [PubMed] [Google Scholar]
- Kirkland Henry P, Davis M, Howell LL. Effects of cocaine self-administration history under limited and extended access conditions on in vivo striatal dopamine neurochemistry and acoustic startle in rhesus monkeys. Psychopharmacol. 2009;205:237–247. doi: 10.1007/s00213-009-1534-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kittappa R, Chang WW, Awatramani RB, McKay RD. The foxa2 gene controls the birth and spontaneous degeneration of dopamine neurons in old age. PLoS Biol. 2007;5:e325. doi: 10.1371/journal.pbio.0050325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacol. 2010;35:217–238. doi: 10.1038/npp.2009.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koya E, Golden SA, Harvey BK, Guez-Barber DH, Berkow A, Simmons DE, Bossert JM, Nair SG, Uejima JL, Marin MT, Mitchell TB, Farquhar D, Ghosh SC, Mattson BJ, Hope BT. Targeted disruption of cocaine-activated nucleus accumbens neurons prevents context-specific sensitization. Nat Neurosci. 2009;12:1069–1073. doi: 10.1038/nn.2364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar A, Choi KH, Renthal W, Tsankova NM, Theobald DE, Truong HT, Russo SJ, Laplant Q, Sasaki TS, Whistler KN, Neve RL, Self DW, Nestler EJ. Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron. 2005;48:303–314. doi: 10.1016/j.neuron.2005.09.023. [DOI] [PubMed] [Google Scholar]
- Letchworth SR, Daunais JB, Hedgecock AA, Porrino LJ. Effects of chronic cocaine administration on dopamine transporter mRNA and protein in the rat. Brain Res. 1997;750:214–222. doi: 10.1016/s0006-8993(96)01384-4. [DOI] [PubMed] [Google Scholar]
- Letchworth SR, Sexton T, Childers SR, Vrana KE, Vaughan RA, Davies HM, Porrino LJ. Regulation of rat dopamine transporter mRNA and protein by chronic cocaine administration. J Neurochem. 1999;73:1982–1989. [PubMed] [Google Scholar]
- Li J, Witten DM, Johnstone IM, Tibshirani R. Normalization, testing, and false discovery rate estimation for RNA-sequencing data. Biostatistics. 2012;13:523–538. doi: 10.1093/biostatistics/kxr031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin W, Metzakopian E, Mavromatakis YE, Gao N, Balaskas N, Sasaki H, Briscoe J, Whitsett JA, Goulding M, Kaestner KH, Ang SL. Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development. Dev Biol. 2009;333:386–396. doi: 10.1016/j.ydbio.2009.07.006. [DOI] [PubMed] [Google Scholar]
- Little KY, McLaughlin DP, Zhang L, McFinton PR, Dalack GW, Cook EH, Jr, Cassin BJ, Watson SJ. Brain dopamine transporter messenger RNA and binding sites in cocaine users: A postmortem study. Arch Gen Psychiatry. 1998;55:793–799. doi: 10.1001/archpsyc.55.9.793. [DOI] [PubMed] [Google Scholar]
- Luscher C, Malenka RC. Drug-evoked synaptic plasticity in addiction: From molecular changes to circuit remodeling. Neuron. 2011;69:650–663. doi: 10.1016/j.neuron.2011.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin LJ, Cork LC. The non-human primate striatum undergoes marked prolonged remodeling during postnatal development. Front Cell Neurosci. 2014;8:294. doi: 10.3389/fncel.2014.00294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mash DC, ffrench-Mullen J, Adi N, Qin Y, Buck A, Pablo J. Gene expression in human hippocampus from cocaine abusers identifies genes which regulate extracellular matrix remodeling. PloS one. 2007;2:e1187. doi: 10.1371/journal.pone.0001187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mavromatakis YE, Lin W, Metzakopian E, Ferri AL, Yan CH, Sasaki H, Whisett J, Ang SL. Foxa1 and Foxa2 positively and negatively regulate Shh signalling to specify ventral midbrain progenitor identity. Mech Dev. 2011;128:90–103. doi: 10.1016/j.mod.2010.11.002. [DOI] [PubMed] [Google Scholar]
- Maze I, Feng J, Wilkinson MB, Sun H, Shen L, Nestler EJ. Cocaine dynamically regulates heterochromatin and repetitive element unsilencing in nucleus accumbens. Proc Nat Acad Sci USA. 2011;108:3035–3040. doi: 10.1073/pnas.1015483108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meredith GE, Baldo BA, Andrezjewski ME, Kelley AE. The structural basis for mapping behavior onto the ventral striatum and its subdivisions. Brain Struct Funct. 2008;213:17–27. doi: 10.1007/s00429-008-0175-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meredith GE, Pattiselanno A, Groenewegen HJ, Haber SN. Shell and core in monkey and human nucleus accumbens identified with antibodies to calbindin-D28k. J Compar Neurol. 1996;365:628–639. doi: 10.1002/(SICI)1096-9861(19960219)365:4<628::AID-CNE9>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
- Metzakopian E, Bouhali K, Alvarez-Saavedra M, Whitsett JA, Picketts DJ, Ang SL. Genome-wide characterisation of Foxa1 binding sites reveals several mechanisms for regulating neuronal differentiation in midbrain dopamine cells. Development. 2015;142:1315–1324. doi: 10.1242/dev.115808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Metzakopian E, Lin W, Salmon-Divon M, Dvinge H, Andersson E, Ericson J, Perlmann T, Whitsett JA, Bertone P, Ang SL. Genome-wide characterization of Foxa2 targets reveals upregulation of floor plate genes and repression of ventrolateral genes in midbrain dopaminergic progenitors. Development. 2012;139:2625–2634. doi: 10.1242/dev.081034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer M, Briggs AW, Maricic T, Hober B, Hoffner B, Krause J, Weihmann A, Paabo S, Hofreiter M. From micrograms to picograms: Quantitative PCR reduces the material demands of high-throughput sequencing. Nucleic Acids Res. 2008;36:e5. doi: 10.1093/nar/gkm1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirabella AC, Foster BM, Bartke T. Chromatin deregulation in disease. Chromosoma. 2016;125:75–93. doi: 10.1007/s00412-015-0530-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakatani T, Kumai M, Mizuhara E, Minaki Y, Ono Y. Lmx1a and Lmx1b cooperate with Foxa2 to coordinate the specification of dopaminergic neurons and control of floor plate cell differentiation in the developing mesencephalon. Dev Biol. 2010;339:101–113. doi: 10.1016/j.ydbio.2009.12.017. [DOI] [PubMed] [Google Scholar]
- Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005;8:1445–1449. doi: 10.1038/nn1578. [DOI] [PubMed] [Google Scholar]
- Nestler EJ. Epigenetic mechanisms of drug addiction. Neuropharmacol. 2014;76(Pt B):259–268. doi: 10.1016/j.neuropharm.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paxinos G, Huang X-F, Toga AW. The Rhesus Monkey Brain In Stereotaxic Coordinates 2000 [Google Scholar]
- Piechota M, Korostynski M, Solecki W, Gieryk A, Slezak M, Bilecki W, Ziolkowska B, Kostrzewa E, Cymerman I, Swiech L, Jaworski J, Przewlocki R. The dissection of transcriptional modules regulated by various drugs of abuse in the mouse striatum. Genome Biol. 2010;11:R48. doi: 10.1186/gb-2010-11-5-r48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Platt DM, Carey G, Spealman RD. Models of neurological disease (substance abuse): Self-administration in monkeys. In: Enna SJ, editor. Current protocols in pharmacology / editorial board. Unit10. Chapter 10. 2011. p. 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Platt DM, Rowlett JK. Nonhuman primates in drug and alcohol addiction research. In: Abee C, Mansfield K, Morris T, Tardif S, editors. Nonhuman Primates in Biomedical Research. Elsevier Press; Waltham, MA: 2012. pp. 817–839. [Google Scholar]
- Renthal W, Maze I, Krishnan V, Covington HE, 3rd, Xiao G, Kumar A, Russo SJ, Graham A, Tsankova N, Kippin TE, Kerstetter KA, Neve RL, Haggarty SJ, McKinsey TA, Bassel-Duby R, Olson EN, Nestler EJ. Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli. Neuron. 2007;56:517–529. doi: 10.1016/j.neuron.2007.09.032. [DOI] [PubMed] [Google Scholar]
- Rodriguez-Espinosa N, Fernandez-Espejo E. Effects of acute and repeated cocaine on markers for neural plasticity within the mesolimbic system in rats. Psychopharmacol. 2015;232:57–62. doi: 10.1007/s00213-014-3632-0. [DOI] [PubMed] [Google Scholar]
- Saddoris MP, Sugam JA, Cacciapaglia F, Carelli RM. Rapid dopamine dynamics in the accumbens core and shell: learning and action. Front Biosci. 2013;5:273–288. doi: 10.2741/e615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sadri-Vakili G. Cocaine triggers epigenetic alterations in the corticostriatal circuit. Brain Res. 2014;1628:50–59. doi: 10.1016/j.brainres.2014.09.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt HD, McGinty JF, West AE, Sadri-Vakili G. Epigenetics and psychostimulant addiction. Cold Spring Harb Perspect Med. 2013;3:a012047. doi: 10.1101/cshperspect.a012047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt SV, Krebs W, Ulas T, Xue J, Bassler K, Gunther P, Hardt AL, Schultze H, Sander J, Klee K, Theis H, Kraut M, Beyer M, Schultze JL. The transcriptional regulator network of human inflammatory macrophages is defined by open chromatin. Cell Res. 2016;26:151–170. doi: 10.1038/cr.2016.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schroeder A, Mueller O, Stocker S, Salowsky R, Leiber M, Gassmann M, Lightfoot S, Menzel W, Granzow M, Ragg T. The RIN: An RNA integrity number for assigning integrity values to RNA measurements. BMC Mol Biol. 2006;7:3. doi: 10.1186/1471-2199-7-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proc Nat Acad Sci USA. 2003;100:9440–9445. doi: 10.1073/pnas.1530509100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stott SR, Metzakopian E, Lin W, Kaestner KH, Hen R, Ang SL. Foxa1 and foxa2 are required for the maintenance of dopaminergic properties in ventral midbrain neurons at late embryonic stages. J Neurosci. 2013;33:8022–8034. doi: 10.1523/JNEUROSCI.4774-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang WX, Fasulo WH, Mash DC, Hemby SE. Molecular profiling of midbrain dopamine regions in cocaine overdose victims. J Neurochem. 2003;85:911–924. doi: 10.1046/j.1471-4159.2003.01740.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL, Pachter L. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protocols. 2012;7:562–578. doi: 10.1038/nprot.2012.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vrana SL, Vrana KE, Koves TR, Smith JE, Dworkin SI. Chronic cocaine administration increases CNS tyrosine hydroxylase enzyme activity and mRNA levels and tryptophan hydroxylase enzyme activity levels. J Neurochem. 1993;61:2262–2268. doi: 10.1111/j.1471-4159.1993.tb07468.x. [DOI] [PubMed] [Google Scholar]
- Weerts EM, Fantegrossi WE, Goodwin AK. The value of nonhuman primates in drug abuse research. Exp Clin Psychopharmacol. 2007;15:309–327. doi: 10.1037/1064-1297.15.4.309. [DOI] [PubMed] [Google Scholar]
- Yuferov V, Kroslak T, Laforge KS, Zhou Y, Ho A, Kreek MJ. Differential gene expression in the rat caudate putamen after “binge” cocaine administration: Advantage of triplicate microarray analysis. Synapse. 2003;48:157–169. doi: 10.1002/syn.10198. [DOI] [PubMed] [Google Scholar]
- Zhang D, Zhang L, Tang Y, Zhang Q, Lou D, Sharp FR, Zhang J, Xu M. Repeated cocaine administration induces gene expression changes through the dopamine D1 receptors. Neuropsychopharmacol. 2005;30:1443–1454. doi: 10.1038/sj.npp.1300680. [DOI] [PubMed] [Google Scholar]
- Zhang JS, Wang L, Huang H, Nelson M, Smith DI. Keratin 23 (K23), a novel acidic keratin, is highly induced by histone deacetylase inhibitors during differentiation of pancreatic cancer cells. Genes Chromosomes Cancer. 2001;30:123–135. [PubMed] [Google Scholar]
- Zhang X, Goodsell J, Norgren RB., Jr Limitations of the rhesus macaque draft genome assembly and annotation. BMC Genomics. 2012;13:206. doi: 10.1186/1471-2164-13-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Michelhaugh SK, Schmidt CJ, Liu JS, Bannon MJ, Lin Z. Ventral midbrain correlation between genetic variation and expression of the dopamine transporter gene in cocaine-abusing versus non-abusing subjects. Addict Biol. 2014a;19:122–131. doi: 10.1111/j.1369-1600.2011.00391.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Z, Enoch MA, Goldman D. Gene expression in the addicted brain. Int Rev Neurobiol. 2014b;116:251–273. doi: 10.1016/B978-0-12-801105-8.00010-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Z, Yuan Q, Mash DC, Goldman D. Substance-specific and shared transcription and epigenetic changes in the human hippocampus chronically exposed to cocaine and alcohol. Proc Nat Acad Sci USA. 2011;108:6626–6631. doi: 10.1073/pnas.1018514108. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.