Abstract
Background
Paclitaxel is an important chemotherapeutic agent for the treatment of breast cancer. Paclitaxel-induced peripheral neuropathy (PIPN) is a major dose-limiting toxicity that can persist into survivorship. While not all survivors develop PIPN, for those who do, it has a substantial negative impact on their functional status and quality of life. No interventions are available to treat PIPN. In our previous studies, we identified that the HIF-1 signaling pathway (H1SP) was perturbed between breast cancer survivors with and without PIPN. Preclinical studies suggest that the H1SP is involved in the development of bortezomib-induced and diabetic peripheral neuropathy, and sciatic nerve injury. The purpose of this study was to identify H1SP genes that have both differential methylation and differential gene expression between breast cancer survivors with and without PIPN.
Methods
A multi-staged integrated analysis was performed. In peripheral blood, methylation was assayed using microarray and gene expression was assayed using RNA-seq. Candidate genes in the H1SP having both differentially methylation and differential expression were identified between survivors who received paclitaxel and did (n = 25) and did not (n = 25) develop PIPN. Then, candidate genes were evaluated for differential methylation and differential expression in public data sets of preclinical models of PIPN and sciatic nerve injury.
Results
Eight candidate genes were identified as both differential methylation and differential expression in survivors. Of the eight homologs identified, one was found to be differential expression in both PIPN and “normal” mice dorsal root ganglia; three were differential methylation in sciatic nerve injury versus sham rats in both pre-frontal cortex and T-cells; and two were differential methylation in sciatic nerve injury versus sham rats in the pre-frontal cortex.
Conclusions
This study is the first to evaluate for methylation in cancer survivors with chronic PIPN. The findings provide evidence that the expression of H1SP genes associated with chronic PIPN in cancer survivors may be regulated by epigenetic mechanisms and suggests genes for validation as potential therapeutic targets.
Keywords: Paclitaxel-induced peripheral neuropathy, breast cancer, survivor, chemotherapy, gene expression, methylation, hypoxia-inducible factor, integrated genomic analysis
Introduction
Paclitaxel is an important chemotherapeutic agent for the treatment of breast cancer.1 Peripheral neuropathy is an adverse effect that occurs in 59% to 87% of patients who receive paclitaxel2,3 and can persist into survivorship.4 Paclitaxel-induced peripheral neuropathy (PIPN) has a substantial negative effect on survivors’ functional status and quality of life.5–11
Not all patients develop this neurotoxicity, which supports the suggestion that molecular mechanisms may be involved in the development of PIPN. Because research with preclinical models of neuropathic pain have not yielded effective treatments, studies that use samples from patients and survivors with chemotherapy-induced peripheral neuropathy (CIPN)12,13 are needed to address this gap. In our previous studies that evaluated gene expression patterns in survivors with and without persistent PIPN,14–16 we identified perturbed pathways associated with mitochondrial dysfunction,14 neuroinflamation,15 and changes in cytoskeleton and axon morphology.16 These findings are consistent with mechanisms identified in preclinical models of neuropathic pain including CIPN.17–23
One of the common pathways across these three mechanisms is the hypoxia-inducible factor 1 (HIF-1) signaling pathway. This pathway, a master regulator of cellular responses to hypoxia,24 plays an important role in axon regeneration following peripheral nerve injury.25 HIF-α is a transcription factor (TF) and global regulator of oxygen homeostasis that facilitates oxygen delivery and adaptation to oxygen deprivation.26,27 It mediates processes that protect against axonal degeneration28 and stimulate axon regeneration.29 While our recent studies were the first to report on an association between PIPN and the HIF-1 pathway,14–16 findings from preclinical studies suggest that this TF is involved in mechanisms that underlie the development of bortezomib-induced peripheral neuropathy,30 diabetic peripheral neuropathy,31 and sciatic nerve injury.32
Given that our previous studies of PIPN in breast cancer survivors provide evidence for differential gene expression and pathway perturbations14–16 and that epigenetics modifications may be involved in the development of neuropathic pain33,34 as well as involved in the transition from acute to chronic pain,35,36 we sought to evaluate for epigenetic mechanisms37 that may influence the expression of these products.38 One potential epigenetic mechanism is DNA methylation, which regulates gene expression by adding or removing methyl groups at the 5′-position of DNA cytosine residues.39 Methylation of DNA can impact transcription by recruiting or blocking TFs, which results in changes in gene expression.40 Epigenetic variations can occur in response to environmental factors (e.g., CTX).38 The identification of genes with both differential methylation (DM) and differential expression patterns associated with PIPN may provide useful information on loci that exhibit changes in function and regulation.
In a recent review,41 it was noted that opportunities may exist to develop or repurpose existing drugs that target the epigenome42 to treat a variety of neuropathic pain conditions including PIPN. Therefore, an increased understanding of DM in biologic pathways associated with the occurrence of PIPN may provide new insights into how these pathways are regulated and suggest targets for drug development. Given the role that epigenetics may play in the development and maintenance of neuropathic pain,33,35 the purpose of this study was to evaluate for differentially methylated and differentially expressed genes in the HIF-1 signaling pathway in breast cancer survivors with (n = 25) and without (n = 25) chronic PIPN and evaluate these candidates in pre-existing data sets from animal models of neuropathic pain including PIPN.
Methods
Survivors and settings
The methods for this analysis, which is part of a larger study of CIPN, are described in detail elsewhere.11 In brief, survivors were recruited from throughout the San Francisco Bay area and met pre-specified inclusion and exclusion criteria. The National Coalition for Cancer Survivorship’s definition of cancer survivor was used in this study (i.e., a person is a cancer survivor from the moment of diagnosis through the balance of life).43 Of the 1450 survivors who were screened, 754 enrolled, and 623 completed the self-report questionnaires and study visit. Data from a randomly selected sample of breast cancer survivors with (n = 25) or without (n = 25) chronic PIPN were used in this analysis.
Study procedures
Research nurses screened and consented the survivors over the phone; sent and asked them to complete the self-report questionnaires prior to their study visit; and scheduled the in-person assessment. At this assessment, written informed consent was obtained, responses to questionnaires were reviewed for completeness, and objective measurements were obtained. Blood samples were drawn, processed, and stored for subsequent molecular analyses in PAXgene® Blood RNA tubes (Qiagen, Venlo, the Netherlands). This study was approved by the institutional review board of the University of California, San Francisco.
Study measures
Demographic and clinical characteristics
Breast cancer survivors provided information on demographic characteristics and completed the Alcohol Use Disorders Identification Test (AUDIT),44 Karnofsky Performance Status (KPS) scale,45–47 and Self-Administered Comorbidity Questionnaire (SCQ).48,49
Pain measures
Survivors with PIPN rated their pain intensity using a 0 to 10 numeric rating scale and completed the pain interference scale from the Brief Pain Inventory50 and the Pain Quality Assessment Scale.51
Biospecimen processing, quantification of methylation status, and quality control
DNA samples from buffy coats were archived using the PUREGene DNA isolation kit (Invitrogen, Carlsbad, CA) as previously described.52,53 DNA samples were quantitated with a NanoDrop UV spectrophotometer (Thermo Fisher Scientific, Waltham, MA) and normalized to a concentration of 50 ng/μL. The DNA preparation and microarray work were performed at the UC Berkeley Vincent J. Coates Genomics Sequencing Laboratory. DNA was bisulfite converted using the Zymo EZ-96 DNA Methylation Kit (Catalog #D5004) Deep-Well Format (Zymo Research, Irvine, CA) and used as input for the Illumina Infinium HD Methylation Assay (Illumina, San Diego, CA). Processed DNA was dispensed onto the Infinium MethylationEPIC BeadChip and scanned on the Illumina iScan (Illumina). Preliminary analysis and quality control (QC) of the data were performed using GenomeStudio (Illumina). Target success rates were determined. Samples that had <90% of their targets methylated with a detection p-value of ≤0.01 were flagged for review. Sample replicates and Jurkat control replicates were checked to ensure an r2 value of >0.99.
Subsequent data analyses were done using well-established protocols in R (version 3.6.1).54,55 Corrections for Infinium I and II probes, balance correction, background correction, and quantile normalization were performed using the minfi package in R (version 1.30.0).56,57 Probes that contained a single nucleotide polymorphism at a CpG or flanking site and probes that aligned to multiple places on the genome were excluded.58 Methylation scores were quantified as M-values.59 Probes were annotated for genes based on mapping to the Genome Reference Consortium Human Build 38 (GRCh38) assembly.60
Because DNA methylation differs among blood cell types,61–63 cell types were estimated using the estimateCellCounts2() function in the FlowSorted.Blood.EPIC R package (version 1.2.0).64 Cell type deconvolution was performed using the IDOL L-DMR library for CD8 and CD4 T-cells, natural killer (NK) cells, B cells, monocytes, and neutrophils. Differences between PIPN groups in estimates of cell type composition were evaluated using t-tests and assessed for significance at a p-value of <0.05. Any cell type composition estimates that were significantly associated with PIPN group membership were included as covariates in the final model.
Surrogate variable analysis (SVA, R package version 3.32.1)65,66 was used to identify technical variations that contributed to heterogeneity in the sample (e.g., batch effects) that were not due to the variable of interest (i.e., PIPN), significant demographic (i.e., age, employment status) and clinical (i.e., AUDIT score, body mass index (BMI), KPS score) characteristics.66 SVA can control for cell type heterogeneity in the methylation analysis.62,63 The “be” method was used to identify surrogate variables.65,66 Two surrogate variables associated with RIN, extraction date, and cell type composition estimates were included in the analysis.67
Overlapping DM and gene expression
To understand the association of PIPN and biological variation in HIF-1 pathway genes, we performed a multi-staged integrated analysis using complementary layers of molecular data.68,69 In the first stage, DM analysis of probes between cases and non-cases was performed using the limma R package (version 3.40.6).70 A linear model was fit using the “ls” method that included significant demographic (i.e., age, employment status) and clinical (i.e., AUDIT score, BMI, KPS score) characteristics, significant cell type composition associated with PIPN (i.e., CD4T cells), and surrogate variables as covariates in the models. We estimates that a minimum of 25 replicates in each group would provide at least 80% power to detect a mean difference in M-values of ≥0.09 at a nominal single locus threshold (type I error rate of 0.01).71
In the second stage, differential gene expression (DGE) was evaluated for each candidate gene using a generalized linear model with edgeR72 as previously described.14 Briefly, these DGE analyses were adjusted for demographic (i.e., age, employment status) and clinical (i.e., AUDIT score, BMI, KPS score) characteristics that differed between the PIPN groups, as well as for technical variability (e.g., potential batch effects) using SVA. We estimated14 that, at a type I error rate of 0.01,73 we were powered to detect 1.5-fold changes for 83% of genes across the whole transcriptome.
Overlapping gene expression and methylation probe pairs were identified at the gene level using the annotated Human Genome Organisation (HUGO) approved symbols using the HUGO Gene Nomenclature Committee (HGNC) database.74 By using a systems genomics design integrating data from multiple biological sources, we would have increased power to identify and better interpret the omics-phenotype relationships relative to an analysis that used only a single source of omics data.69,75 For this exploratory study, candidate genes were defined by a significant overlap assessed at a p-value <0.05 for both DM and expression tests. No minimal fold-change was utilized.
Functional analyses of candidate genes
To characterize potential functional roles and potential interactions among candidate genes with differentially methylated probes and DGE beyond the HIF-1 signaling pathway, we evaluated for protein–protein interaction (PPI) network connectivity using the Search Tool for the Retrieval of Interacting Genes (STRING).76 The functional enrichment of the PPI was evaluated using the Kyoto Encyclopedia of Genes and Genomes (KEGG)77 and Reactome78 pathway databases. We assessed for significance of functional enrichment tests using a false discovery rate of 5 under the Benjamini–Hochberg (BH) procedure.79,80 In addition, we evaluated for similar expression patterns of our candidate genes in various cell types in previously identified peripheral neuroimmune interactions81 from RNA-seq data sets using the “NIPPY—Neuro-Immune Interactions in the Periphery” database (https://rna-seq-browser.herokuapp.com/).
Differential expression of our candidate genes in DRG of mice with and without PIPN
Differential expression of our candidate genes was evaluated using a publicly available data set generated from mouse dorsal root ganglia (DRG) (NCBI GEO GSE113941). Data were collected from nine pools of mice 10 days after treatment with paclitaxel (n = 5) in a dose that was sufficient to induce PIPN (i.e. “CIPN model”) compared to not treated mice (n = 4) (i.e., “normal”). For our study, FASTQ files of the RNA-seq data from these experiments were downloaded from the NCBI Short Read Archive (GSM3124261, GSM3124262, GSM3124263, GSM3124264, GSM3124269, GSM3124270, GSM3124271, GSM3124272, and GSM3124273).
Following our previously described protocols,14 these sequences were trimmed with Trimmomatic and aligned to the mouse genome assembly (GRCm38.p6) annotated by GENCODE vM23 with the STAR aligner.82 Gene level counts were generated using the featureCounts tool in the subread R package (version 2.0.0).83 QC and DGE analyses were performed as previously described.14 The “be” method was used to identify surrogate variables.65,66 No outlier samples were identified and all samples were included in subsequent analyses. Genes with ≤1 read per million in at least four samples were excluded. Two surrogate variables were identified. Neither surrogate variable was associated with neuropathy and both were used as covariates in the final model. DGE was evaluated between the mouse DRG sample pools who did (n = 5) and did not (n = 4) received paclitaxel. These findings were compared to the differentially methylated and expressed candidate genes identified in our cancer survivors. Homologous mouse gene Ensemble ID was determined by the HUGO Symbol using the HGNC Database.74 Significant DGE was assessed at a type I error rate of 0.05. No minimal fold-change was evaluated.
DM of our candidate genes in pre-frontal cortex and T-cells of rats with and without SNI
DM of our candidate genes was evaluated in two publicly available data sets generated from rat pre-frontal cortex (PFC) in sham (n = 8) vs. sciatic nerve injury (SNI)84 (n = 7) animals (GEO GSE70006) and from T-cells in sham (n = 8) vs. SNI (n = 8) animals (GEO GSE70007) that were measured on a customized array (Aglient Technologies, Santa Clara, CA) nine months after the procedure. The PFC and T-cells were harvested from the same animals. Methylation levels were downloaded from GEO as the quantile normalized log2 ratio of the bound (Cy5) and input (Cy3) microarray channel intensities. DM in both experiments was evaluated using limma. Homologous rat genes were identified from their HUGO symbols using the HGNC Database74 and Rat Genome Database.85 Significant DM was assessed at a type I error rate of 0.05. No minimal fold-change was utilized.
Results
Differences in demographic, clinical, and pain characteristics
Cancer survivor characteristics were reported previously.14 In brief, breast cancer survivors with chronic PIPN were significantly older (p = 0.006) and were more likely to be unemployed (p = 0.022) (Supplemental Table 1). In terms of clinical characteristics, survivors with PIPN had a lower AUDIT score (p = 0.012), a higher BMI (p = 0.011), and a lower KPS score (p < 0.001) (Supplemental Table 2). Of note, no between-group differences were found in the number of years since cancer diagnosis, the total dose of paclitaxel received or in the percentage of patients who had a dose reduction or delay due to PIPN. Worst pain severity was 6.3 (±2.1) and duration of PIPN was 3.8 (±3.9) years (Supplemental Table 3).
DM and DGE associated with PIPN in survivors
For the 100 genes that were identified in the KEGG HIF-1 signaling pathway, we mapped 732 probes that had a regulatory feature group classified as “promoter associated” and evaluated for DM. Only CD4+ T-cell composition estimates were associated with PIPN group membership (Supplemental Table 4). Eleven surrogate variables were identified. One was associated with PIPN group membership and four were identified as significantly associated with cell type composition. The final regression model included five significant demographic and clinical characteristics (i.e., age, employment status, AUDIT score, BMI, KPS score) and four surrogate variables. We had methylation and expression data for 81 genes in the HIF-1 signaling pathway that were candidates for evaluation for both DGE and DM. Twelve probes across eight genes were identified as both differentially methylated and expressed between survivors with and without PIPN (Table 1).
Table 1.
Genes in the HIF-1 pathway that were differentially methylated at promoter associated sites and differentially expressed between breast cancer survivors with and without paclitaxel-induced peripheral neuropathy.
HGNC-approved symbol | HGNC-approved name | HGNC Alias symbols | Ensemble Gene ID | logFC (Gx) | p-value (Gx) | Probe ID | logFC (Mt) | p-value (Mt) |
---|---|---|---|---|---|---|---|---|
TFRC | Transferrin receptor | CD71, TFR1, p90 | ENSG00000072274 | −0.863 | 1.95 × 10−5 | cg27335386 | −0.011 | 0.012 |
cg26126750 | 0.025 | 0.013 | ||||||
cg14641705 | −0.014 | 0.021 | ||||||
cg25254330 | 0.006 | 0.039 | ||||||
RBX1 | Ring-box 1 | BA554C12.1, RNF75, ROC1 | ENSG00000100387 | −0.762 | 1.91 × 10−4 | cg10781403 | −0.015 | 0.031 |
cg19850545 | −0.011 | 0.038 | ||||||
PFKL | Phosphofructokinase, liver type | – | ENSG00000141959 | 0.295 | 0.012 | cg04665109 | 0.020 | 0.009 |
CUL2 | Cullin 2 | – | ENSG00000108094 | −0.292 | 0.012 | cg27370344 | −0.007 | 0.043 |
MKNK1 | MAPK interacting serine/threonine kinase 1 | MNK1 | ENSG00000079277 | −0.235 | 0.015 | cg03409548 | 0.043 | 0.037 |
EGLN1 | Egl-9 family HIF 1 | C1orf12, HIFPH2, PHD2, SM-20, ZMYND6 | ENSG00000135766 | −0.286 | 0.019 | cg21365899 | 0.007 | 0.026 |
LDHA | Lactate dehydrogenase A | – | ENSG00000134333 | −0.009 | 0.037 | cg03177631 | −0.009 | 0.037 |
MAP2K2 | MAPK kinase 2 | PRKMK2, MEK2 | ENSG00000126934 | −0.295 | 0.045 | cg03016975 | 0.035 | 0.014 |
Gx: gene expression; HIF: hypoxia-inducible factor; HGNC: HUGO Gene Nomenclature Committee; HUGO: Human Genome Organisation; logFC: log2 fold change; MAPK: mitogen-activated protein kinase; Mt: methylation.
Functional analysis identified seven KEGG and three Reactome pathways that were enriched for the eight genes listed in Table 1 (excluding the HIF-1 signaling pathway itself; Table 2). These eight genes represent a significantly enriched PPI network (p = 0.002) and the network identified evidence of protein–protein interactions between six genes (Figure 1).
Table 2.
Functionally enriched pathways associated from eight genes in the HIF-1 signaling pathway with overlapping differential methylation and differential expression between breast cancer survivors with and without paclitaxel-induced peripheral neuropathy.
ID | Name | Source | FDR | Nset | Npathway |
---|---|---|---|---|---|
hsa04066 | HIF-1 signaling pathway | KEGG | 5.0e-17 | 8 | 98 |
hsa05211 | Renal cell carcinoma | KEGG | 5.2e-07 | 4 | 68 |
hsa05230 | Central carbon metabolism in cancer | KEGG | 6.6e-05 | 3 | 65 |
hsa05200 | Pathways in cancer | KEGG | 7.0e-04 | 4 | 515 |
R-HSA-1234176 | Oxygen-dependent proline hydroxylation of HIFA | Reactome | 2.5e-04 | 3 | 62 |
hsa00010 | Glycolysis/Gluconeogenesis | KEGG | 6.2e-03 | 2 | 68 |
hsa04922 | Glucagon signaling pathway | KEGG | 0.01 | 2 | 100 |
hsa04120 | Ubiquitin mediated proteolysis | KEGG | 0.02 | 2 | 134 |
hsa04910 | Insulin signaling pathway | KEGG | 0.02 | 2 | 134 |
R-HSA-422475 | Axon guidance | Reactome | 0.02 | 3 | 541 |
R-HSA-9010553 | Regulation of expression of SLITs and ROBOs | Reactome | 0.04 | 2 | 164 |
FDR: false discovery rate; FGF: fibroblast growth factor; HIF: hypoxia-inducible factor; HIFA: HIF alpha; KEGG: Kyoto Encyclopedia of Genes and Genomes; MAP: mitogen-activated protein kinase; Nset: count of candidate genes of the query set found in the pathway; Npathway: count of genes in the pathway; RAF: rapidly accelerated fibrosarcoma; RUNX2: runt-related transcription factor 2; TGF: transforming growth factor.
Figure 1.
STRING connectivity network analysis identified protein–protein interactions between cullin 2 (CUL2), egl-9 family hypoxia-inducible factor 1 (EGLN1), ring-box 1 (RBX1), lactate dehydrogenase A (LDHA), transferrin receptor (TFRC), and phosphofructokinase, liver type (PFKL). Nodes represent all proteins produced by a single protein coding gene locus. Edges represent specific or meaningful associations. Known or predicted 3D structures are presented within the nodes. Color of the edges connecting the nodes represents the types of evidence supporting the connections: predicted gene neighborhood (green), predicted gene fusions (red), known interactions from experimental evidence (pink), co-expression (black), and text-mining (green).
Differential expression of candidate genes in mouse DRG associated with PIPN
Of the eight candidate genes identified as both differentially methylated and expressed in breast cancer survivors, eight homologs were identified in the mouse DRG data set (Table 3). Of these eight candidates, one gene (i.e., Mknk1) was found to be differentially expressed between PIPN and “normal” pools of mice DRG.
Table 3.
Test for differential expression of candidate genes in the HIF-1 pathway in DRG between pools of mice treated with paclitaxel and normal controls.
H.s. Symbol | M.m. Symbol | Description | MGIAccession(s) | NCBI Gene | Ensemble GeneID | logFC | p-value |
---|---|---|---|---|---|---|---|
TFRC | Tfrc | Transferrin receptor | 9882 | 22042 | ENSMUSG00000022797 | −0.03 | 0.861 |
RBX1 | Rbx1 | Ring-box 1 | 1891829 | 56438 | ENSMUSG00000022400 | −0.28 | 0.111 |
PFKL | Pfkl | Phosphofructokinase, liver, B-type | 97547 | 18641 | ENSMUSG00000020277 | −0.30 | 0.293 |
CUL2 | Cul2 | Cullin 2 | 1918995 | 71745 | ENSMUSG00000024231 | −0.09 | 0.622 |
MKNK1 | Mknk1 | MAPK-interactingSerine/threonine kinase 1 | 894316 | 17346 | ENSMUSG00000028708 | 1.12 | 0.005 |
EGLN1 | Egln1 | egl-9 family HIF 1 | 1932286 | 112405 | ENSMUSG00000031987 | 0.24 | 0.238 |
LDHA | Ldha | Lactate dehydrogenase A | 96759 | 16828 | ENSMUSG00000063229 | −0.38 | 0.256 |
MAP2K2 | Mapk2k2 | mitogen-activated protein kinase kinase 2 | 1346867 | 26396 | ENSMUSG00000035027 | 0.28 | 0.168 |
DRG: dorsal root ganglia; HIF: hypoxia-inducible factor; H.s.: Homo sapiens; logFC: log2 fold change; MAPK: mitogen-activated protein kinase; MGI: Mouse Genome Informatics; M.m.: Mus musculus; NCBI: National Center for Biotechnology Information.
DM of candidate genes in rat PFC and T-cells associated with SNI
Of the eight candidate genes with overlapping DM and DGE in breast cancer survivors, seven homologs were identified in the rat PFC and T-cell data set (i.e., Mknk1, Map2k2, Egln1, Rbx1, Pfkl, Cul2, Ldha). For these seven genes, we identified 72 probes (Egln1 n = 14, Map2k2 n = 10, Mknk1 n = 8, Pfkl n = 12, Rbx1 n = 5, Cul2 n = 12, Ldha n = 11) in the promoter regions and evaluated them for DM. Of these seven candidates, three genes (i.e., Mknk1, Ldha, Egln1) were differentially methylated between SNI and sham rats in both PFC and T-cells (Table 4). While no genes were differentially methylated only in T-cells, two genes (i.e., Cul2, Map2k2) were differentially methylated only in the PFC.
Table 4.
Differential methylation of probes in promoter regions of candidate genes in pre-frontal cortex and T-cells of rats with spared nerve injury versus sham.
R.n. Symbol | RGD ID | Description | Spot ID | Tissue Type | logFC | p-value |
---|---|---|---|---|---|---|
Cul2 | 1310644 | Cullin 2 | ||||
NM_001108417:-898 | PFC | 1.42 | 0.013 | |||
NM_001108417:-890 | PFC | 0.90 | 0.024 | |||
NM_001108417:-820 | PFC | 1.83 | 0.023 | |||
Egln1 | 631375 | Egl-9 family hypoxia-inducible factor 1 | ||||
NM_178334:-1542 | T-cell | −1.20 | 0.043 | |||
NM_178334:-1514 | PFC | 1.18 | 0.017 | |||
NM_178334:-1424 | PFC | 1.96 | 0.002 | |||
Ldha | 2996 | Lactate dehydrogenase A | ||||
NM_017025:-1018 | T-cell | −1.67 | 0.015 | |||
NM_017025:-645 | PFC | 1.62 | 0.025 | |||
NM_017025:-44 | PFC | −0.64 | 0.013 | |||
Map2k2 | 61888 | Mitogen-activated protein kinase kinase 2 | ||||
NM_133283:-117 | PFC | −0.76 | 0.002 | |||
Mknk1 | 1559603 | MAPK-interacting serine/threonine kinase 1 | ||||
NM_178334:-1424 | PFC | 1.97 | 0.002 | |||
NM_001044267:-854 | T-cell | −1.77 | 0.015 | |||
NM_001044267:-19 | T-cell | −1.28 | 0.028 |
logFC: log2 Fold Change; MAPK: mitogen-activated protein kinase; PFC: pre-frontal cortex; R.n.: Rattus norvegicus; RGD: Rat Genome Database; Spot ID: GenBank Locus:position.
Discussion
This study is the first to use a multi-staged integrated analysis to identify genes in the HIF-1 signaling pathway that were both differentially methylated and differentially expressed in breast cancer survivors with PIPN. In addition, this study is the first to identify a subset of these genes as being differentially methylated or differentially expressed in preclinical models of PIPN86 or SNI.84 Pre-clinical animal models are needed to increase our understanding of the fundamental biological mechanisms that underlie neuropathic.87–89 Given that these types of experiments cannot be done in humans, a comparison of our findings in cancer survivors with those from pre-clinical models allows us to gain insights into their translatability. Of note, one of these HIF-1 signaling pathway candidate genes (i.e., MKNK1/Mknk1) was associated with both DGE in mouse DRG and DM in rat PFC and T-cells. The remainder of this discussion focuses on these genes and evaluates their potential role in the mechanisms that underlie PIPN and the implications of these findings with respect to epigenetic regulation of gene expression.
HIF-1 signaling pathway genes associated with DM and DGE
The mitogen-activated protein kinase (MAPK) 1 interacting serine/threonine kinase 1 (MKNK1) gene is involved in the control of the cellular proteome90 and in inflammatory responses.41 MAPK interreacting protein kinases (Mnks) are broadly expressed across different tissues types. In terms of neuroinflammation, Mnks play critical roles in cytokine receptor signaling.41 MKNK1 mediates cytokine production in macrophages through post-transcriptional regulation and can mediate TNF-α translation.41
In terms of effecting translation, MKNK1 phosphorylates the eukaryotic initiation factor 4E (eIF4E) and is activated downstream of the p38 or MAPK pathways.41 The regulation of the eIF4E complex is a key signaling pathway that controls the cellular proteome.91 We found that the MKNK1 gene was differentially methylated and expressed in peripheral blood from breast cancer survivors with PIPN, differentially methylated in PFC and T-cells of rats with SNI, and differentially expressed in DRG of mice with PIPN. Recent work suggests that changes in Mknk1 expression and phosphorylation of elF4E in mice contribute to nociceptor plasticity92–94 and that inhibition or elimination of Mkn1 (Mknk1) attenuates PIPN in a murine model.93 In addition, inhibition of Mkn1 and Mnk2 reduces spontaneous pain in a SNI murine model.95 Interestingly, the phosphorylation of elF4E by Mknk1 was identified in a neurological model animal, Aplysia californica, which suggests that this regulatory mechanism for neuroplasticity is highly conserved96 across Bilateria.97
In terms of neuroinflammation, Mknk1 is expressed in both DRG and peripheral blood and across peripheral and neuroimmune cell lines (Supplemental Figures 1 and 2) which is consistent with our findings of expression across tissue types. Compared to survivors without PIPN, MKNK1 had lower expression and higher methylation in survivors with PIPN. In contrast to our findings in survivors, Mknk1 had higher expression in mice with PIPN compared to normal mice. One possible explanation for this difference is the timing of the measures. For the mice with PIPN, gene expression was measured 10 days after CTX compared to approximately four years in our cancer survivors (Supplemental Table 2). Similarly, in a recent preclinical study81 of traumatic nerve injury, findings from macrophage and T-cells dissected from DRG suggests that Mknk1 expression levels decline 10 weeks after sciatic nerve injury (Supplemental Figure 4). Based on the fact that inflammation persisted unresolved for 3.5 months (the duration of the study) in these animals, the authors argued that the categorization of pain as either inflammatory or neuropathic may not be mechanistically appropriate. Given that DNA methylation reprogramming was observed in preclinical models of chronic neuropathic pain following nerve injury,36 future research should evaluate for changes in the methylation and expression levels of Mknk1 over time.
MAP2K2 is a kinase which phosphorylates and activates MAPK1 and MAPK3.98 The MAPK1 gene is involved in eukaryotic signal transduction99 and is associated with the development of neuropathic pain.100,101 We found that MAP2K2/Map2k2 was differentially methylated and differentially expressed in peripheral blood from breast cancer survivors with PIPN and differentially methylated in PFC of rats with SNI. ERK2 (MAPK1) activation by the three MAPK family pathways (i.e., phosphoinositide 3-kinases (PI3Ks), adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR)) promotes inflammatory and neuropathic pain conditions (reviewed in Wood et al.102). Evaluations of the effects of paclitaxel on cancer cell lines found that paclitaxel activated these MAPK family pathways.103–105 Thus, the regulation of MAPK1 was an early therapeutic target for the treatment of neuropathic pain induced by peripheral nerve injury.106 More recently, inhibition of a number of MAPK family pathways (e.g., p38, MAPK, and JNK) was shown to attenuate neuropathic and inflammatory pain in preclinical models (reviewed in Ji et al.101). While no studies have examined associations between PIPN and changes in gene expression or methylation of MAP2K2, expression of Map2k2 increased in rat L4 and L5 DRGs cells with sciatic nerve lesions as compared with sham controls three days after injury.107
The transferrin receptor (TFRC) gene encodes for a protein important for cellular iron uptake and homeostatis.108 TFRC (TFR1) is a key iron metabolism gene that fine tunes cellular iron access, storage, and utilization.109 Iron overload can lead to increased oxidative stress110 and may be regulated by HIF-1 TFs.111 We found that TFRC was differentially methylated and expressed in peripheral blood of breast cancer survivors with PIPN. However, data were not available to test for DM in T-cells or PFC of rats with SNI or DGE in DRG of mice treated with paclitaxel. While no studies have examined associations between PIPN and changes in gene expression or methylation of TFRC, polymorphisms in TFRC decreased the risk for distal neuropathic pain in HIV-infected patients on combination antiretroviral therapy.112 In addition, iron overload is a risk factor for diabetic neuropathy.113 In a preclinical central pain model, mRNA expression of TFR1 (an alias for TFRC) was upregulated in activated microglia after spinal cord injury.114 Finally, our previous finding of perturbations in iron homeostasis pathways (i.e., Ferroptosis) in this sample14 suggests that future studies should investigate the role of TFRC in the regulation of iron homeostasis in survivors with PIPN.
The phosphofructokinase, liver type (PFKL) gene codes for the liver subtype of an enzyme that catalyzes the phosphorylation of D-fructose 6-phosphate to fructose 1,6-bisphosphate. Therefore, PFKL is a key metabolic gene in glycolysis.115 We found that PFKL was differentially methylated and expressed in peripheral blood from breast cancer survivors with PIPN. However, it was not DM in T-cells or PFC of rats with SNI or differentially expressed in DRG of mice treated with paclitaxel. No studies were found that evaluated for associations between gene expression or methylation of PFKL and CIPN. However, in a preclinical model of diabetic neuropathy,116 PFKL was induced in IMS32 Schwann cells following chronic treatment with high levels of glucose (>8 weeks). Given that oncology patients with diabetes are at increased risk for developing CIPN,117 future studies should evaluate this mechanism in patients with both comorbidities.
The lactate dehydrogenase A (LDHA) gene catalyzes the forward and backward conversion of lactate to pyruvate in glycolysis. LDHA is activated by HIF118,119 and the reduction of LDHA can induce oxidative stress.120 We found that LDHA was differentially methylated and expressed in peripheral blood of breast cancer survivors with PIPN and differentially methylated in both T-cells and PFC of rats with SNI. Although no studies were found on associations between gene expression or methylation of LDHA and PIPN, LDHA-driven aerobic glycolysis was associated with pain in bortezomib-induced CIPN.121 Given that we found an enrichment in the glycolysis/gluconeogenesis KEGG pathway for these eight candidate genes, future research should investigate the role of LDHA in oxidative stress and cellular respiration in survivors with PIPN.
The Egl nine homolog 1 (EGLN1) gene produces a protein which acts as a cellular oxygen sensor that catalyzes the formation of 4-hydroxyproline in HIF alpha proteins and hydroxylates HIF1A and HIF2A. Then, hydroxylated HIFs are degraded through a ubiquination complex. The ring box 1 (RBX1) and cullin 2 (CUL2) genes encode for proteins that act as an E3 ubuquitin-protein ligase, which are involved in mediating the ubiquitination and degradation of proteins. We found that EGLN1/Egln1 was differentially methylated and expressed in peripheral blood of breast cancer survivors with PIPN and differentially methylated in both T-cells and PFC of rats with SNI. In addition, RBX1 and CUL2 were differentially methylated and expressed in peripheral blood of breast cancer survivors with PIPN. However, although Cul2 was differentially methylated in PFC of rats with SNI, we did not find DM of Rbx1 in T-cells or PFC of rats with SNI. In addition, we did not find Rbx1 or Cul2 to be DGE in DRG of mice treated with paclitaxel. In a pharmacological network-based analysis of drug-induced peripheral neuropathy (including Paclitaxel),122 Cul2 was identified as a highly connected significant intermediator between drugs and their pharmacological targets. Although no studies were found on associations between gene expression or methylation of RBX1 and neuropathy, in oxaliplatin-treated embryonic kidney cells, the overexpression of PHD2 (EGLN1) inhibits cold-induced hTRPA1 activation.123 Future studies should evaluate this mechanism in patients having received taxanes, platinums, or both.
Epigenetic regulation of transcription
Translational control is an essential component in the regulation of gene expression124 and may be a core mechanism for the development and maintenance of persistent pain.125 Translational control of molecular processes associated with neuropathy allows for localized translation outside of the cell body in the axons126,127 (i.e., activity-dependent regulation of mRNA translation128) including the regulation of the eIF4E complex by MNKN1. However, the Mnks are effectors of other biological processes (i.e., receptor tyrosine kinase activity, TNF-α mRNA translation, arachidonate release).90
Alternative splicing is an important step in post-transcriptional regulation of gene expression129 and the diversity of various transcript isoforms of a gene are associated with environmental perturbations.130 Alternative splicing is known to occur in the nucleus either during (co-transcriptional splicing) or after transcription (post transcriptional splicing)131 and DNA methylation can play a role in the regulation of alternative splicing.132,133 Recent work describing variations in splicing across immune cells found that the distribution of isoforms was on/off, which suggests switch-like regulatory control.134 Although not currently identified in CIPN, in a model of pre-diabetic polyneuropathy, errors in splicing factors were associated with neuronal dysfunction.135 In addition, alternative splicing appears to play an important role in the development and function of the nervous system,136 including axon guidance.137 In terms of PIPN, two isoforms of the MKNK1 gene can occur through alternative splicing (i.e., MNK1a and MNK1b). However, the MKN1b isoform is not activated by either p38 MAPK or MAPK1.138,139 This finding suggests that the regulation of MKN1b occurs through other mechanisms.41 Given that gene expression can be modulated at multiple levels, from chromatin folding to mRNA translation, future research should continue to evaluate the relative contributions of epigenetic regulation of translation to the development and maintenance of PIPN.
Conclusions
Several limitations warrant consideration. While our sample size was relatively small, we have an extremely well-characterized sample of breast cancer survivors with and without PIPN. While the integration of data from multiple sources we have increased the power to identify omics-phenotype relationships and enabled a more sophisticated interpretation of the findings,69,75 future research with larger sample sizes may improve the resolution of the methylation and gene expression signals. Our findings warrant validation in an independent sample. Of note, no differences were found in the total cumulative dose of paclitaxel that the two groups of survivors received. Given that methylation and gene expression are not independent processes, our findings must be verified in other samples and with other neurotoxic drugs. The utility of peripheral blood as a biomarker or surrogate for neuronal tissue13,140 or as a direct signal (e.g., peripheral neuro-immune interactions81) is still an active area of research.141 Future research should evaluate for peripheral neuro-immune interactions as well as for the utility of peripheral blood as a surrogate for neuronal tissue.
Our findings suggest that the expression of HIF-1 signaling pathway genes associated with chronic PIPN in cancer survivors may be regulated by epigenetic mechanisms. Future studies need to evaluate for epigenetic changes associated with gene expression and alternative splicing in other pathways associated with PIPN, other CTX drugs, and other forms of neuropathy.
Supplementary Material
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding
The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was funded by the National Cancer Institute (NCI, CA151692) and the American Cancer Society (ACS, IRG-97–150-13). Dr. Miaskowski is supported by grants from the ACS and NCI (CA168960). Dr. Olshen is partially supported by the NCI Cancer Center Support Grant to UCSF (CA082103). This project was also supported by the National Center for Advancing Translational Sciences, National Institutes of Health, through UCSF-CTSI Grant Number UL1 TR000004. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH. Recruitment was facilitated by Dr. Susan Love Research Foundation’s Army of Women® Program.
ORCID iD
Kord M Kober https://orcid.org/0000-0001-9732-3321
Supplemental Material
Supplemental material for this article is available online.
References
- 1.Kudlowitz D, Muggia F. Defining risks of taxane neuropathy: insights from randomized clinical trials. Clin Cancer Res 2013; 19: 4570–4577. [DOI] [PubMed] [Google Scholar]
- 2.Sarosy G, Kohn E, Stone DA, Rothenberg M, Jacob J, Adamo DO, Ognibene FP, Cunnion RE, Reed E, Cunnion RE, Reed E. Phase I study of taxol and granulocyte colony-stimulating factor in patients with refractory ovarian cancer. J Clin Oncol 1992; 10: 1165–1170. [DOI] [PubMed] [Google Scholar]
- 3.Jones SE, Erban J, Overmoyer B, Budd GT, Hutchins L, Lower E, Laufman L, Sundaram S, Urba WJ, Pritchard KI, Mennel R, Richards D, Olsen S, Meyers ML, Ravdin PM. Randomized phase III study of docetaxel compared with paclitaxel in metastatic breast cancer. JCO 2005; 23: 5542–5551. [DOI] [PubMed] [Google Scholar]
- 4.Banach M, Juranek JK, Zygulska AL. Chemotherapy-induced neuropathies-a growing problem for patients and health care providers. Brain Behav 2017; 7: e00558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Beijers AJ, Mols F, Tjan-Heijnen VC, Faber CG, van de Poll-Franse LV, Vreugdenhil G. Peripheral neuropathy in colorectal cancer survivors: the influence of oxaliplatin administration. Results from the population-based PROFILES registry. Acta Oncol 2015; 54: 463–469. [DOI] [PubMed] [Google Scholar]
- 6.Tofthagen C, Donovan KA, Morgan MA, Shibata D, Yeh Y. Oxaliplatin-induced peripheral neuropathy’s effects on health-related quality of life of colorectal cancer survivors. Support Care Cancer 2013; 21: 3307–3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tofthagen C, Visovsky C, Berry DL. Strength and balance training for adults with peripheral neuropathy and high risk of fall: current evidence and implications for future research. Oncol Nurs Forum 2012; 39: E416–E424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ezendam NP, Pijlman B, Bhugwandass C, Pruijt JF, Mols F, Vos MC, Pijnenborg JM, van de Poll-Franse LV. Chemotherapy-induced peripheral neuropathy and its impact on health-related quality of life among ovarian cancer survivors: results from the population-based PROFILES registry. Gynecol Oncol 2014; 135: 510–517. [DOI] [PubMed] [Google Scholar]
- 9.Gewandter JS, Fan L, Magnuson A, Mustian K, Peppone L, Heckler C, Hopkins J, Tejani M, Morrow GR, Mohile SG. Falls and functional impairments in cancer survivors with chemotherapy-induced peripheral neuropathy (CIPN): a University of Rochester CCOP study. Support Care Cancer 2013; 21: 2059–2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Miaskowski C, Mastick J, Paul SM, Abrams G, Cheung S, Sabes JH, Kober KM, Schumacher M, Conley YP, Topp K, Smoot B, Mausisa G, Mazor M, Wallhagen M, Levine JD. Impact of chemotherapy-induced neurotoxicities on adult cancer survivors’ symptom burden and quality of life. J Cancer Surviv. 2018; 12: 234–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Miaskowski C, Mastick J, Paul SM, Topp K, Smoot B, Abrams G, Chen LM, Kober KM, Conley YP, Chesney M, Bolla K, Mausisa G, Mazor M, Wong M, Schumacher M, Levine JD. Chemotherapy-induced neuropathy in cancer survivors. J Pain Symptom Manage 2017; 54: 204–218.e202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.North RY, Li Y, Ray P, Rhines LD, Tatsui CE, Rao G, Johansson CA, Zhang H, Kim YH, Zhang B, Dussor G, Kim TH, Price TJ, Dougherty PM. Electrophysiological and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain 2019; 142: 1215–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bell JT, Loomis AK, Butcher LM, Gao F, Zhang B, Hyde CL, Sun J, Wu H, Ward K, Harris J, Scollen S, Davies MN, Schalkwyk LC, Mill J, Mu TC, Williams FM, Li N, Deloukas P, Beck S, McMahon SB, Wang J, John SL, Spector TD, MuTHER Consortium. Differential methylation of the TRPA1 promoter in pain sensitivity. Nat Commun 2014; 5: 2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kober KM, Olshen A, Conley YP, Schumacher M, Topp K, Smoot B, Mazor M, Chesney M, Hammer M, Paul SM, Levine JD, Miaskowski C. Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain 2018; 14: 1744806918816462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Miaskowski C, Topp K, Conley YP, Paul SM, Melisko M, Schumacher M, Chesney M, Abrams G, Levine JD, Kober KM. Perturbations in neuroinflammatory pathways are associated with paclitaxel-induced peripheral neuropathy in breast cancer survivors. J Neuroimmunol 2019; 335: 577019–577008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kober KM, Schumacher M, Conley YP, Topp K, Mazor M, Hammer MJ, Paul SM, Levine JD, Miaskowski C. Signaling pathways and gene co-expression modules associated with cytoskeleton and axon morphology in breast cancer survivors with chronic paclitaxel-induced peripheral neuropathy. Mol Pain 2019; 15: 1744806919878088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ma J, Kavelaars A, Dougherty PM, Heijnen CJ. Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: targeting the source. Cancer 2018; 124: 2289–2298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Waseem M, Kaushik P, Tabassum H, Parvez S. Role of mitochondrial mechanism in chemotherapy-induced peripheral neuropathy. Curr Drug Metab 2018; 19: 47–54. [DOI] [PubMed] [Google Scholar]
- 19.Flatters SJL, Dougherty PM, Colvin LA. Clinical and preclinical perspectives on chemotherapy-induced peripheral neuropathy (CIPN): a narrative review. Br J Anaesth 2017; 119: 737–749. [DOI] [PubMed] [Google Scholar]
- 20.Makker PG, Duffy SS, Lees JG, Perera CJ, Tonkin RS, Butovsky O, Park SB, Goldstein D, Moalem-Taylor G. Characterisation of immune and neuroinflammatory changes associated with chemotherapy-induced peripheral neuropathy. PLoS One 2017; 12: e0170814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.LaPointe NE, Morfini G, Brady ST, Feinstein SC, Wilson L, Jordan MA. Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: implications for chemotherapy-induced peripheral neuropathy. Neurotoxicology 2013; 37: 231–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shemesh OA, Spira ME. Paclitaxel induces axonal microtubules polar reconfiguration and impaired organelle transport: implications for the pathogenesis of paclitaxel-induced polyneuropathy. Acta Neuropathol 2010; 119: 235–248. [DOI] [PubMed] [Google Scholar]
- 23.Fukuda Y, Li Y, Segal RA. A mechanistic understanding of axon degeneration in chemotherapy-induced peripheral neuropathy. Front Neurosci 2017; 11: 481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zepeda AB, Pessoa A, Jr, Castillo RL, Figueroa CA, Pulgar VM, Farias JG. Cellular and molecular mechanisms in the hypoxic tissue: role of HIF-1 and ROS. Cell Biochem Funct 2013; 31: 451–459. [DOI] [PubMed] [Google Scholar]
- 25.Venkatesh I, Blackmore MG. Selecting optimal combinations of transcription factors to promote axon regeneration: why mechanisms matter. Neurosci Lett 2017; 652: 64–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Semenza GL. Oxygen homeostasis. Wiley Interdiscip Rev Syst Biol Med 2010; 2: 336–361. [DOI] [PubMed] [Google Scholar]
- 27.Semenza GL. Hypoxia-inducible factor 1: regulator of mitochondrial metabolism and mediator of ischemic preconditioning. Biochim Biophys Acta 2011; 1813: 1263–1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Keswani SC, Bosch-Marcé M, Reed N, Fischer A, Semenza GL, Höke A. Nitric oxide prevents axonal degeneration by inducing HIF-1-dependent expression of erythropoietin. Proc Natl Acad Sci USA 2011; 108: 4986–4990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cho Y, Shin JE, Ewan EE, Oh YM, Pita-Thomas W, Cavalli V. Activating injury-responsive genes with hypoxia enhances axon regeneration through neuronal HIF-1alpha. Neuron 2015; 88: 720–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ludman T, Melemedjian OK. Bortezomib and metformin opposingly regulate the expression of hypoxia-inducible factor alpha and the consequent development of chemotherapy-induced painful peripheral neuropathy. Mol Pain 2019; 15: 1744806919850043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Rojas DR, Tegeder I, Kuner R, Agarwal N. Hypoxia-inducible factor 1alpha protects peripheral sensory neurons from diabetic peripheral neuropathy by suppressing accumulation of reactive oxygen species. J Mol Med 2018; 96: 1395–1405. [DOI] [PubMed] [Google Scholar]
- 32.Kanngiesser M, Mair N, Lim HY, Zschiebsch K, Blees J, Haussler A, Brune B, Ferreiros N, Kress M, Tegeder I. Hypoxia-inducible factor 1 regulates heat and cold pain sensitivity and persistence. Antioxid Redox Signal 2014; 20: 2555–2571. [DOI] [PubMed] [Google Scholar]
- 33.Descalzi G, Ikegami D, Ushijima T, Nestler EJ, Zachariou V, Narita M. Epigenetic mechanisms of chronic pain. Trends Neurosci 2015; 38: 237–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Machelska H, Celik MO. Recent advances in understanding neuropathic pain: glia, sex differences, and epigenetics. F1000Res 2016; 5: 2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Buchheit T, Van de Ven T, Shaw A. Epigenetics and the transition from acute to chronic pain. Pain Med 2012; 13: 1474–1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Garriga J, Laumet G, Chen SR, Zhang Y, Madzo J, Issa JJ, Pan HL, Jelinek J. Nerve injury-induced chronic pain is associated with persistent DNA methylation reprogramming in dorsal root ganglion. J Neurosci 2018; 38: 6090–6101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Razin A, Riggs AD. DNA methylation and gene function. Science 1980; 210: 604–610. [DOI] [PubMed] [Google Scholar]
- 38.Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003; 33 Suppl: 245–254. [DOI] [PubMed] [Google Scholar]
- 39.Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity (Edinb) 2010; 105: 4–13. [DOI] [PubMed] [Google Scholar]
- 40.Stephens KE, Miaskowski CA, Levine JD, Pullinger CR, Aouizerat BE. Epigenetic regulation and measurement of epigenetic changes. Biol Res Nurs 2013; 15: 373–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Joshi S, Platanias LC. Mnk kinases in cytokine signaling and regulation of cytokine responses. Biomol Concepts 2012; 3: 127–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Sisignano M, Parnham MJ, Geisslinger G. Drug repurposing for the development of novel analgesics. Trends Pharmacol Sci 2016; 37: 172–183. [DOI] [PubMed] [Google Scholar]
- 43.Clark EJ, Teamwork – the cancer patient’s guide to talking with your doctor. 5th ed Silver Springs, MD: National Coalition for Cancer Survivorship, 2011. [Google Scholar]
- 44.Bohn MJ, Babor TF, Kranzler HR. The Alcohol Use Disorders Identification Test (AUDIT): validation of a screening instrument for use in medical settings. J Stud Alcohol 1995; 56: 423–432. [DOI] [PubMed] [Google Scholar]
- 45.Karnofsky D. Performance scale. New York: Plenum Press, 1977. [Google Scholar]
- 46.Karnofsky D, Abelmann WH, Craver LV, Burchenal JH. The use of nitrogen mustards in the palliative treatment of carcinoma. Cancer 1948; 1: 634–656. [Google Scholar]
- 47.Schnadig ID, Fromme EK, Loprinzi CL, Sloan JA, Mori M, Li H, Beer TM. Patient-physician disagreement regarding performance status is associated with worse survivorship in patients with advanced cancer. Cancer 2008; 113: 2205–2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Brunner F, Bachmann LM, Weber U, Kessels AG, Perez RS, Marinus J, Kissling R. Complex regional pain syndrome 1–the Swiss cohort study. BMC Musculoskelet Disord 2008; 9: 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cieza A, Geyh S, Chatterji S, Kostanjsek N, Ustun BT, Stucki G. Identification of candidate categories of the International Classification of Functioning Disability and Health (ICF) for a Generic ICF Core Set based on regression modelling. BMC Med Res Methodol 2006; 6: 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Daut RL, Cleeland CS, Flanery RC. Development of the Wisconsin Brief Pain Questionnaire to assess pain in cancer and other diseases. Pain 1983; 17: 197–210. [DOI] [PubMed] [Google Scholar]
- 51.Jensen MP, Gammaitoni AR, Olaleye DO, Oleka N, Nalamachu SR, Galer BS. The pain quality assessment scale: assessment of pain quality in carpal tunnel syndrome. J Pain 2006; 7: 823–832. [DOI] [PubMed] [Google Scholar]
- 52.Dhruva A, Aouizerat BE, Cooper B, Paul SM, Dodd M, West C, Wara W, Lee K, Dunn LB, Langford DJ, Merriman JD, Baggott C, Cataldo J, Ritchie C, Kober KM, Leutwyler H, Miaskowski C. Cytokine gene associations with self-report ratings of morning and evening fatigue in oncology patients and their family caregivers. Biol Res Nurs 2015; 17: 175–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Aouizerat BE, Dhruva A, Paul SM, Cooper BA, Kober KM, Miaskowski C. Phenotypic and molecular evidence suggests that decrements in morning and evening energy are distinct but related symptoms. J Pain Symptom Manage 2015; 50: 599–614.e593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bock C. Analysing and interpreting DNA methylation data. Nat Rev Genet 2012; 13: 705–719. [DOI] [PubMed] [Google Scholar]
- 55.Flowers E, Flentje A, Levine J, Olshen A, Hammer M, Paul S, Conley Y, Miaskowski C, Kober K. A pilot study using a multi-staged integrated analysis of gene expression and methylation to evaluate mechanisms for evening fatigue. Biol Res Nurs in Nurs 2019; 21: 142–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Du P, Kibbe WA, Lin SM. lumi: a pipeline for processing Illumina microarray. Bioinformatics 2008; 24: 1547–1548. [DOI] [PubMed] [Google Scholar]
- 57.Aryee MJ, Jaffe AE, Corrada-Bravo H, Ladd-Acosta C, Feinberg AP, Hansen KD, Irizarry RA. Minfi: a flexible and comprehensive bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics 2014; 30: 1363–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, Gallinger S, Hudson TJ, Weksberg R. Discovery of cross-reactive probes and polymorphic CpGs in the Illumina Infinium HumanMethylation450 microarray. Epigenetics 2013; 8: 203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Du P, Zhang X, Huang CC, Jafari N, Kibbe WA, Hou L, Lin SM. Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics 2010; 11: 587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhou W, Laird PW, Shen H. Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Res 2017; 45: e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Koestler DC, Christensen B, Karagas MR, Marsit CJ, Langevin SM, Kelsey KT, Wiencke JK, Houseman EA. Blood-based profiles of DNA methylation predict the underlying distribution of cell types: a validation analysis. Epigenetics 2013; 8: 816–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.McGregor K, Bernatsky S, Colmegna I, Hudson M, Pastinen T, Labbe A, Greenwood CM. An evaluation of methods correcting for cell-type heterogeneity in DNA methylation studies. Genome Biol 2016; 17: 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Houseman EA, Molitor J, Marsit CJ. Reference-free cell mixture adjustments in analysis of DNA methylation data. Bioinformatics 2014; 30: 1431–1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Salas LA, Koestler DC, Butler RA, Hansen HM, Wiencke JK, Kelsey KT, Christensen BC. An optimized library for reference-based deconvolution of whole-blood biospecimens assayed using the Illumina HumanMethylationEPIC BeadArray. Genome Biol 2018; 19: 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Leek JT, Johnson WE, Parker HS, Jaffe AEa, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics 2012; 28: 882–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet 2007; 3: 1724–1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Jaffe AE, Hyde T, Kleinman J, Weinbergern DR, Chenoweth JG, McKay RD, Leek JT, Colantuoni C. Practical impacts of genomic data “cleaning” on biological discovery using surrogate variable analysis. BMC Bioinformatics 2015; 16: 372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Buescher JM, Driggers EM. Integration of omics: more than the sum of its parts. Cancer Metab 2016; 4: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ritchie MD, Holzinger ER, Li R, Pendergrass SA, Kim D. Methods of integrating data to uncover genotype-phenotype interactions. Nat Rev Genet 2015; 16: 85–97. [DOI] [PubMed] [Google Scholar]
- 70.Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015; 43: e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Tsai PC, Bell JT. Power and sample size estimation for epigenome-wide association scans to detect differential DNA methylation. Int J Epidemiol 2015; 44: 1429–1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010; 26: 139–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hart SN, Therneau TM, Zhang Y, Poland GA, Kocher JP. Calculating sample size estimates for RNA sequencing data. J Comput Biol 2013; 20: 970–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Yates B, Braschi B, Gray KA, Seal RL, Tweedie S, Bruford EA. Genenames.org: the HGNC and VGNC resources in 2017. Nucleic Acids Res 2017; 45: D619–D625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ideker T, Dutkowski J, Hood L. Boosting signal-to-noise in complex biology: prior knowledge is power. Cell 2011; 144: 860–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von Mering C. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 2015; 43: D447–D452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Aoki-Kinoshita KF, Kanehisa M. Gene annotation and pathway mapping in KEGG. Methods Mol Biol (Clifton, NJ) 2007; 396: 71–91. [DOI] [PubMed] [Google Scholar]
- 78.Joshi-Tope G, Gillespie M, Vastrik I, D'Eustachio P, Schmidt E, de Bono B, Jassal B, Gopinath GR, Wu GR, Matthews L, Lewis S, Birney E, Stein L. Reactome: a knowledgebase of biological pathways. Nucleic Acids Res 2005; 33: D428–D432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hochberg Y, Benjamini Y. More powerful procedures for multiple significance testing. Stat Med 1990; 9: 811–818. [DOI] [PubMed] [Google Scholar]
- 80.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B (Methodol) 1995; 57: 289–300. [Google Scholar]
- 81.Liang Z, Hore Z, Harley P, Stanley FU, Michrowska A, Dahiya M, La Russa F, Jager SE, Villa-Hernandez S, Denk F. A transcriptional toolbox for exploring peripheral neuro-immune interactions. bioRxiv: 813980, 2019, www.biorxiv.org/content/10.1101/813980v1 [DOI] [PubMed] [Google Scholar]
- 82.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29: 15–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014; 30: 923–930. [DOI] [PubMed] [Google Scholar]
- 84.Cichon J, Sun L, Yang G. Spared nerve injury model of neuropathic pain in mice. Bio Protoc 2018; 8: e2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Shimoyama M, De Pons J, Hayman GT, Laulederkind SJ, Liu W, Nigam R, Petri V, Smith JR, Tutaj M, Wang SJ, Worthey E, Dwinell M, Jacob H. The Rat Genome Database 2015: genomic, phenotypic and environmental variations and disease. Nucleic Acids Res 2015; 43: D743–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol 2011; 25: 1–28. [DOI] [PubMed] [Google Scholar]
- 87.Clark JD. Preclinical pain research: can we do better? Anesthesiology 2016; 125: 846–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Burma NE, Leduc-Pessah H, Fan CY, Trang T. Animal models of chronic pain: advances and challenges for clinical translation. J Neurosci Res 2017; 95: 1242–1256. [DOI] [PubMed] [Google Scholar]
- 89.Gregory NS, Harris AL, Robinson CR, Dougherty PM, Fuchs PN, Sluka KA. An overview of animal models of pain: disease models and outcome measures. J Pain 2013; 14: 1255–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Joshi S, Platanias LC. Mnk kinase pathway: cellular functions and biological outcomes. World J Biol Chem 2014; 5: 321–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Richter JD, Sonenberg N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature 2005; 433: 477–480. [DOI] [PubMed] [Google Scholar]
- 92.Gkogkas CG, Khoutorsky A, Cao R, Jafarnejad SM, Prager-Khoutorsky M, Giannakas N, Kaminari A, Fragkouli A, Nader K, Price TJ, Konicek BW, Graff JR, Tzinia AK, Lacaille JC, Sonenberg N. Pharmacogenetic inhibition of eIF4E-dependent Mmp9 mRNA translation reverses fragile X syndrome-like phenotypes. Cell Rep 2014; 9: 1742–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Megat S, Ray PR, Moy JK, Lou TF, Barragan-Iglesias P, Li Y, Pradhan G, Wanghzou A, Ahmad A, Burton MD, North RY, Dougherty PM, Khoutorsky A, Sonenberg N, Webster KR, Dussor G, Campbell ZT, Price TJ. Nociceptor translational profiling reveals the Ragulator-Rag GTPase complex as a critical generator of neuropathic pain. J Neurosci 2019; 39: 393–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Moy JK, Khoutorsky A, Asiedu MN, Dussor G, Price TJ. eIF4E phosphorylation influences Bdnf mRNA translation in mouse dorsal root ganglion neurons. Front Cell Neurosci 2018; 12: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Shiers S, Mwirigi J, Pradhan G, Kume M, Black B, Barragan-Iglesias P, Moy JK, Dussor G, Pancrazio JJ, Kroener S, Price TJ. Reversal of peripheral nerve injury-induced neuropathic pain and cognitive dysfunction via genetic and tomivosertib targeting of MNK. Neuropsychopharmacology 2020; 45: 524–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mihail SM, Wangzhou A, Kunjilwar KK, Moy JK, Dussor G, Walters ET, Price TJ. MNK-eIF4E signalling is a highly conserved mechanism for sensory neuron axonal plasticity: evidence from Aplysia californica. Philos Trans R Soc Lond, B, Biol Sci 2019; 374: 20190289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Valentine JW. Cleavage patterns and the topology of the metazoan tree of life. Proc Natl Acad Sci USA 1997; 94: 8001–8005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Crews CM, Alessandrini A, Erikson RL. The primary structure of MEK, a protein kinase that phosphorylates the ERK gene product. Science 1992; 258: 478–480. [DOI] [PubMed] [Google Scholar]
- 99.Seger R, Krebs EG. The MAPK signaling cascade. Faseb J 1995; 9: 726–735. [PubMed] [Google Scholar]
- 100.Obata K, Noguchi K. MAPK activation in nociceptive neurons and pain hypersensitivity. Life Sci 2004; 74: 2643–2653. [DOI] [PubMed] [Google Scholar]
- 101.Ji R-R, Gereau RW, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev 2009; 60: 135–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wood JN, Willemen H, Eijkelkamp N. Sensory signaling pathways in inflammatory and neuropathic pain. Oxford: Oxford University Press, 2019. [Google Scholar]
- 103.Okano J, Rustgi AK. Paclitaxel induces prolonged activation of the Ras/MEK/ERK pathway independently of activating the programmed cell death machinery. J Biol Chem 2001; 276: 19555–19564. [DOI] [PubMed] [Google Scholar]
- 104.Lieu CH, Liu CC, Yu TH, Chen KD, Chang YN, Lai YK. Role of mitogen-activated protein kinase in taxol-induced apoptosis in human leukemic U937 cells. Cell Growth Differ 1998; 9: 767–776. [PubMed] [Google Scholar]
- 105.McDaid HM, Horwitz SB. Selective potentiation of paclitaxel (taxol)-induced cell death by mitogen-activated protein kinase kinase inhibition in human cancer cell lines. Mol Pharmacol 2001; 60: 290–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ma W, Quirion R. The ERK/MAPK pathway, as a target for the treatment of neuropathic pain. Expert Opin Ther Targets 2005; 9: 699–713. [DOI] [PubMed] [Google Scholar]
- 107.Zhang CG, Wan HQ, Ma KN, Luan SX, Li H. Identification of biomarkers related to neuropathic pain induced by peripheral nerve injury. J Mol Neurosci 2019; 69: 505–515. [DOI] [PubMed] [Google Scholar]
- 108.Gammella E, Buratti P, Cairo G, Recalcati S. The transferrin receptor: the cellular iron gate. Metallomics 2017; 9: 1367–1375. [DOI] [PubMed] [Google Scholar]
- 109.Bogdan AR, Miyazawa M, Hashimoto K, Tsuji Y. Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends Biochem Sci 2016; 41: 274–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Berg D, Youdim MB. Role of iron in neurodegenerative disorders. Top Magn Reson Imaging 2006; 17: 5–17. [DOI] [PubMed] [Google Scholar]
- 111.Peyssonnaux C, Zinkernagel AS, Schuepbach RA, Rankin E, Vaulont S, Haase VH, Nizet V, Johnson RS. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest 2007; 117: 1926–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kallianpur AR, Jia P, Ellis RJ, Zhao Z, Bloss C, Wen W, Marra CM, Hulgan T, Simpson DM, Morgello S, McArthur JC, Clifford DB, Collier AC, Gelman BB, McCutchan JA, Franklin D, Samuels DC, Rosario D, Holzinger E, Murdock DG, Letendre S, Grant I, Group CS, for the CHARTER Study Group. Genetic variation in iron metabolism is associated with neuropathic pain and pain severity in HIV-infected patients on antiretroviral therapy. PLoS One 2014; 9: e103123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhao S, Zhang L, Xu Z, Chen W. Neurotoxic effects of iron overload under high glucose concentration. Neural Regen Res 2013; 8: 3423–3433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Meng FX, Hou JM, Sun TS. In vivo evaluation of microglia activation by intracranial iron overload in central pain after spinal cord injury. J Orthop Surg Res 2017; 12: 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yi W, Clark PM, Mason DE, Keenan MC, Hill C, Goddard WA, Peters EC, Driggers EM, Hsieh-Wilson LC. Phosphofructokinase 1 glycosylation regulates cell growth and metabolism. Science 2012; 337: 975–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kim ES, Isoda F, Kurland I, Mobbs CV. Glucose-induced metabolic memory in Schwann cells: prevention by PPAR agonists. Endocrinology 2013; 154: 3054–3066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hershman DL, Till C, Wright JD, Awad D, Ramsey SD, Barlow WE, Minasian LM, Unger J. Comorbidities and risk of chemotherapy-induced peripheral neuropathy among participants 65 years or older in Southwest Oncology Group clinical trials. JCO 2016; 34: 3014–3022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Semenza GL, Jiang BH, Leung SW, Passantino R, Concordet JP, Maire P, Giallongo A. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J Biol Chem 1996; 271: 32529–32537. [DOI] [PubMed] [Google Scholar]
- 119.Firth JD, Ebert BL, Ratcliffe PJ. Hypoxic regulation of lactate dehydrogenase A. Interaction between hypoxia-inducible factor 1 and cAMP response elements. J Biol Chem 1995; 270: 21021–21027. [DOI] [PubMed] [Google Scholar]
- 120.Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL, Semenza GL, Dang CV. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA 2010; 107: 2037–2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ludman T, Melemedjian OK. Bortezomib-induced aerobic glycolysis contributes to chemotherapy-induced painful peripheral neuropathy. Mol Pain 2019; 15: 1744806919837429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hur J, Guo AY, Loh WY, Feldman EL, Bai JP. Integrated systems pharmacology analysis of clinical drug-induced peripheral neuropathy. CPT Pharmacometrics Syst Pharmacol 2014; 3: e114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Miyake T, Nakamura S, Meng Z, Hamano S, Inoue K, Numata T, Takahashi N, Nagayasu K, Shirakawa H, Mori Y, Nakagawa T, Kaneko S. Distinct mechanism of cysteine oxidation-dependent activation and cold sensitization of human transient receptor potential Ankyrin 1 channel by high and low oxaliplatin. Front Physiol 2017; 8: 878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hershey JW, Sonenberg N, Mathews MB. Principles of translational control: an overview. Cold Spring Harb Perspect Biol. 2019; 11: a032607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Khoutorsky A, Price TJ. Translational control mechanisms in persistent pain. Trends Neurosci 2018; 41: 100–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Wang W, van Niekerk E, Willis DE, Twiss JL. RNA transport and localized protein synthesis in neurological disorders and neural repair. Dev Neurobiol 2007; 67: 1166–1182. [DOI] [PubMed] [Google Scholar]
- 127.Sahoo PK, Smith DS, Perrone-Bizzozero N, Twiss JL. Axonal mRNA transport and translation at a glance. J Cell Sci 2018; 131: jcs196808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Di Liegro CM, Schiera G, Di Liegro I. Regulation of mRNA transport, localization and translation in the nervous system of mammals (Review). Int J Mol Med 2014; 33: 747–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lee Y, Rio DC. Mechanisms and regulation of alternative pre-mRNA splicing. Annu Rev Biochem 2015; 84: 291–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Pai AA, Luca F. Environmental influences on RNA processing: biochemical, molecular and genetic regulators of cellular response. Wiley Interdiscip Rev RNA 2019; 10: e1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Han J, Xiong J, Wang D, Fu XD. Pre-mRNA splicing: where and when in the nucleus. Trends Cell Biol 2011; 21: 336–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lev Maor G, Yearim A, Ast G. The alternative role of DNA methylation in splicing regulation. Trends Genet 2015; 31: 274–280. [DOI] [PubMed] [Google Scholar]
- 133.Luco RF, Allo M, Schor IE, Kornblihtt AR, Misteli T. Epigenetics in alternative pre-mRNA splicing. Cell 2011; 144: 16–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ergun A, Doran G, Costello JC, Paik HH, Collins JJ, Mathis D, Benoist C, ImmGen C, ImmGen Consortium. Differential splicing across immune system lineages. Proc Natl Acad Sci USA 2013; 110: 14324–14329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kobayashi M, Zochodne DW. Diabetic neuropathy and the sensory neuron: new aspects of pathogenesis and their treatment implications. J Diabetes Investig 2018; 9: 1239–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Norris AD, Calarco JA. Emerging roles of alternative pre-mRNA splicing regulation in neuronal development and function. Front Neurosci 2012; 6: 122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Su C-H, D D, Tarn W-Y. Alternative splicing in neurogenesis and brain development. Front Mol Biosci 2018; 5: 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Goto S, Yao Z, Proud CG. The C-terminal domain of Mnk1a plays a dual role in tightly regulating its activity. Biochem J 2009; 423: 279–290. [DOI] [PubMed] [Google Scholar]
- 139.Scheper GC, Parra JL, Wilson M, Van Kollenburg B, Vertegaal AC, Han ZG, Proud CG. The N and C termini of the splice variants of the human mitogen-activated protein kinase-interacting kinase Mnk2 determine activity and localization. Mol Cell Biol 2003; 23: 5692–5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Massart R, Dymov S, Millecamps M, Suderman M, Gregoire S, Koenigs K, Alvarado S, Tajerian M, Stone LS, Szyf M. Overlapping signatures of chronic pain in the DNA methylation landscape of prefrontal cortex and peripheral T cells. Sci Rep 2016; 6: 19615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yu X, Liu H, Hamel KA, Morvan MG, Yu S, Leff J, Guan Z, Braz JM, Basbaum AI. Dorsal root ganglion macrophages contribute to both the initiation and persistence of neuropathic pain. Nat Commun 2020; 11: 264. [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.