Abstract
Epigenetics has gained considerable interest as potential mediators of molecular alterations that could underlie the prolonged sensitization of nociceptors, neurons, and glia in response to various environmental stimuli. Histone acetylation and deacetylation, key processes in modulating chromatin, influence gene expression; elevated histone acetylation enhances transcriptional activity, whereas decreased acetylation leads to DNA condensation and gene repression. Altered levels of histone deacetylase (HDAC) have been detected in various animal pain models, and HDAC inhibitors have demonstrated analgesic effects in these models, indicating HDACs’ involvement in chronic pain pathways. However, animal studies have predominantly examined epigenetic modulation within the spinal cord after pain induction, which may not fully reflect the complexity of chronic pain in humans. Moreover, methodological limitations have previously impeded an in-depth study of epigenetic changes in the human brain. In this study, we employed [11C]Martinostat, an HDAC-selective radiotracer, positron emission tomography to assess HDAC availability in the brains of 23 chronic low back pain (cLBP) patients and 11 age- and sex-matched controls. Our data revealed a significant reduction of [11C]Martinostat binding in several brain regions associated with pain processing in cLBP patients relative to controls, highlighting the promising potential of targeting HDAC modulation as a therapeutic strategy for cLBP.
Keywords: chronic pain, low back pain, positron emission tomography, neuroepigenetics, histone deacetylases
Introduction
Chronic pain is characterized as persistent pain lasting longer than three months which further unfavorably affect the quality of life [8,52]. In the United States, low back pain affects 80% of adults, and 15–20% of LBP sufferers develop chronic LBP (cLBP), causing significant suffering and socio-economic burden [12,32,53]. Although prescribing opioids for pain management is not preferred, moderate-to-severe pain, including cLBP, remains one of the leading causes of opioid prescription [11,29] because nonpharmacologic treatments or nonsteroidal anti-inflammatory drugs have been reported to be less effective than opioids-based options [20]. Moreover, prescription opioids have undesirable side effects, such as constipation, respiratory depression, and the potential for abuse and dependence [4,29,31]. Therefore, identifying alternatives to current treatment options is an urgent clinical need that can be achieved by advancing our comprehension of pathophysiological mechanisms underlying chronic pain.
Nociceptors within the peripheral nervous system (PNS) and neurons and glial cells in the central nervous system (CNS) undergo a process of sensitization in response to environmental stimuli like tissue damage, inflammation, and infection [27,54]. These stimuli provoke morphological/functional changes based on sustained molecular alterations associated with prolonged pain [16]. Such changes are often amplified by self-perpetuating mechanisms that modulate gene expression [16]. Within this context, the field of epigenetics, which includes DNA methylation, histone acetylation, and non-coding RNA interactions, has become increasingly relevant due to its role in regulating gene expression without altering the underlying DNA sequence. Histone acetylation and deacetylation, as determine by the interplays between histone acetyltransferases and histone deacetylases (HDACs), govern the extent of chromatin relaxation. Elevated histone acetylation enhances transcriptional activity, while reduced acetylation leads to DNA condensation and gene repression. Different levels of HDAC expression and histone acetylation have been observed across various animal models of pain, reflecting dynamic epigenetic regulation [6,9,16,17,28,33,49]. The analgesic effects of HDAC inhibitors in these models [2,9,14] underscore the relevance of HDACs in neuroplastic changes and highlight their potential as therapeutic targets for pain management [9,15,16,25,40]. However, research on histone modifications in response to pain within the CNS is still in its early stages [6], with most research to date focusing on the spinal cord in animal models. It is important to note that these animal models may not fully recapitulate chronic pain in humans. Methodological limitations have impeded explorations of epigenetic changes in the human brain.
Recently, the availability of HDACs can be quantified using an HDAC-selective radiotracer, [11C]Martinostat, with positron emission tomography (PET). [11C]Martinostat exhibits selectivity for a subset of class I/IIb HDACs (class I: HDAC 1, 2, 3 and class IIb: HDAC 6) [39,48,50,51]. [11C]Martinostat showed high binding and appropriate characteristics for imaging, as shown in our previous studies [18,50]. Importantly, the regional distribution of [11C]Martinostat binding aligned well with their relative ranking of HDAC expression levels as measured in post-mortem tissue samples [18,50]. This study aims to explore potential alterations in HDAC availability within the brains of cLBP patients relative to age- and sex-matched healthy controls. Post hoc analyses further examine associations between altered HDAC levels and patient-reported outcomes.
Methods
Study participants
Twenty-three cLBP patients (11 males and 12 females; age: 46.0 ± 14.7 years old) and 11 healthy controls (8 males and 3 females; age: 48.5 ± 17.6 years old) were included in this study. Participants provided written informed consent, which was approved by the Institutional Review Board and the Radioactive Drug Research Committee at Massachusetts General Hospital, to take part in the study. Study data were collected and managed using REDCap (Research Electronic Data Capture) electronic data capture tools hosted at Massachusetts General Hospital [21,22]. Healthy controls were matched to cLBP patients by age (within two years), sex, race, and exclusion criteria, except they have no documentation of low back pain via medical records. Potential cLBP subjects were excluded if they experienced psychiatric impairment that could prevent completion of study procedures, had an active substance use disorder based on DSM-V criteria, had a history of myocardial infarction or other cardiovascular conditions, underwent surgery within six months of contact, tested positive on a urine drug screen (with the exception for cannabis), experienced peripheral neuropathy, migraines, or fibromyalgia, or were contraindicated for magnetic resonance imaging (MRI) or PET scanning. For their pain management, nine opioid-naive cLBP patients reported the use of NSAID medications at the time of screening, and three cLBP patients reported the use of gabapentin for nerve control and back pain, but no subjects reported the use of opioid-based analgesics, which is confirmed by a negative urine drug test. In addition to the above criteria, opioid-naive cLBP patients were excluded if they did not experience moderate-to-severe pain (i.e., above 4/10 on a visual analog scale), if they reported back pain resulting from cancer or bodily injury, had extensive back surgery that would complicate any interpretation of their data, underwent interventional procedures for back pain within two weeks of contact, or could not provide documentation of low back pain via medical records or a note from a clinician.
Each cLBP subject underwent a comprehensive medical history, physical examination, and low back pain evaluation by a medical doctor or nurse practitioner during enrollment. The clinician inquired about the approximate date of pain onset and duration, as well as any associated injuries or acute trauma. Each cLBP subject should have at least 6 months of pain duration. The examination characterized each subject’s pain as axial, pseudo-radicular, or radicular, and determined affected dermatome (if radicular or pseudo-radicular) or location. Current subjective pain levels (0 = “no pain” and 10 = “the most intense pain imaginable”) were also assessed. The clinician reviewed the results with the study team to ensure that no exclusion criteria were met. All cLBP participants also completed a set of questionnaires and psychological assessments, including the Brief Pain Inventory (BPI; for pain severity), the Oswestry Disability Index (ODI; for self-reported function and disability), the Hospital Anxiety Depression Scale (HADS total score; for emotional function)[35], painDETECT (for neuropathic pain elements), the Screener and Opioid Assessment for Patients with Pain, Revised (SOAPP-R; for estimating the risk of opioid misuse)[5], and the Mini International Neuropsychiatric Interview (MINI; to rule out current substance abuse disorder). Additionally, each participant completed the “minimum dataset” suggested by the NIH Back Pain Consortium (BACPAC)[34], as part of the NIH HEAL Initiative, consisting of two questionnaires (Baseline Demographics and Outcomes Assessment) concerning demographics, pain level, pain interference, sleep disturbance, physical function, and emotional function. Healthy controls did not complete assessments on pain but did complete surveys on demographics.
Radiosynthesis of [11C]Martinostat
[11C]Martinostat is a hydroxamic acid-based HDAC inhibitor that contains an adamantyl group and radiolabeled with 11C. Briefly, the synthesis of [11C]Martinostat was initiated by reductive amination, followed by conversion into a hydroxamic acid in the presence of hydroxylamine and sodium hydroxide in accordance with cGMP guidelines as previously described [48,50].
Scan procedures: PET/MR imaging
All participants had no MRI or PET contraindications to undergo brain imaging safely. PET/MRI scans were acquired on a 3T Siemens TIM Trio with a BrainPET insert (Siemens, Erlangen, Germany). A PET-compatible circularly polarized transmit coil, and an eight-channel receive array coil were used for MRI data acquisition. An intravenous catheter was placed in the antecubital vein of the other arm, and a licensed nuclear medicine technologist performed a manual bolus injection (9.23 ± 4.37 mCi) of [11C]Martinostat into the intravenous catheter. Dynamic PET scan (90 min) was started concomitantly with radiotracer injection. Our previous study with healthy individuals reported that image-based outcome measures, i.e., standard uptake values (SUV) and SUV normalized to the white matter (SUVR), are well correlated with the volume of distribution values (VT) derived from kinetic modeling with low inter-subject variability [50]. It is important to confirm whether a simplified outcome measure is valid in cLBP patients. Therefore, in a subset of cLBP patients (n=11), an arterial line was placed in the radial artery of one arm, and blood samples were collected from the arterial line by an experienced nurse practitioner. Participants were instructed to remain still during the scan. Due to a long scan duration, we registered the MR-based Attenuation Correction (AC) maps to preliminary PET recons spanning 60–90 mins after radiotracer administration and carefully performed motion corrections for the reconstructed PET images. Images were reconstructed using the three-dimensional ordinary Poisson ordered-subset expectation maximization (3D OP-OSEM) algorithm with detector efficiency, decay, dead time, attenuation, and scatter corrections applied. An atlas-based MR AC method using Statistical Parametric Mapping (SPM) was applied[24] to derive a linear attenuation coefficient map. The final PET images with the units of SUV (mean radioactivity per injected dose per weight) and radioactivity concentrations (becquerels per milliliter) were reconstructed into 153 slices with 256 × 256 pixels 1.25-mm isotropic voxel size. A high-resolution structural scan using a multi-echo magnetization-prepared rapid acquisition gradient-echo sequence (MEMPRAGE) was acquired during the PET scan using the following parameters: repetition time (TR) = 2530 ms; echo time 1 (TE1) = 1.64 ms; TE2 = 3.49 ms; TE3 = 5.35 ms; TE4 = 7.21 ms; inversion time (TI) = 1200 ms; flip angle = 7°; and isotropic resolution = 1 mm. List mode PET data were stored for 90 min and binned into 30 frames of progressively longer duration (10 s × 9, 20 s × 3, 30 s × 3, 60 s × 1, 120 s × 1, 180 s × 1, 300 s × 8, and 600 s × 4).
Arterial Blood Sampling and Analysis
The arterial blood samples were acquired during the dynamic PET scans at ~10 s intervals beginning at the start of the scan for 3 min (~2 ml each), which was followed by additional samples at 5, 10, 20, 30, 50, 70, and 90 min (~6 ml each) after radiotracer injection for plasma and radiometabolite analyses. In brief, the collected blood samples were centrifuged. The resulting 200 μl (for the samples obtained within the first 3 min) or 600 μl (for other samples) of plasma samples were placed in a gamma counter cross-calibrated with the PET scanner. For the radiometabolite high-performance liquid chromatography (HPLC) analysis, the samples obtained at (5, 10, 20, 30, 50, 70, and 90 min) were centrifuged at 4,000 rpm at 4 °C, for 4 mins. The centrifuged plasma (1 ml) was extracted from the sample tube and added to another tube containing 1 mL of acetonitrile. The resulting tube was briefly vortexed to mix the contents and then it was centrifuged for a second time with the same parameters. The liquid contents (~1 ml) of that tube was extracted and mixed in a scintillation vial containing 4 mL of deionized water with 0.1% Formic Acid. The mixture was gently shaken, and the total 5mL solution was injected on the HPLC (Agilent 1260 HPLC with Fraction Collector, Agilent Technologies, CA, USA). Total plasma radioactivity was linearly interpolated and corrected for the fractions of radiometabolites over time. The metabolite-corrected arterial plasma input curves were used for kinetic modeling (see below).
MR image processing and analysis
Each subject’s high-resolution T1-weighted images were analyzed with automatic segmentation and parcellation using FreeSurfer (version 6.0; http://surfer.nmr.mgh.harvard.edu/). From the segmentation outcomes, subject-specific binary masks for a set of region-of-interests (ROIs) containing a total of 27 brain regions were obtained for the region-based analysis. The set of ROIs includes whole brain, white matter, deep white matter, pons, cerebellum, brain stem, thalamus, caudate, putamen, pallidum, hippocampus, amygdala, nucleus accumbens, periaqueductal gray (PAG), orbitofrontal cortex (OFC), anterior cingulate cortex (ACC), posterior cingulate cortex (PCC), insula, precentral gyrus (primary motor cortex), postcentral gyrus (somatosensory cortex), paracentral lobule, rostral-middle-frontal gyrus (dorsolateral prefrontal cortex; dlFPC), precuneus, inferior-frontal gyrus, supramarginal gyrus (somatosensory association cortex), middle-temporal gyrus, superior-parietal gyrus. The selection of this set of ROIs was driven by two objectives: 1) whole brain, white matter, deep white matter, pons and cerebellum were chosen as potential normalization references for image analysis; 2) other brain regions were selected because they were implicated in the processing of pain, as indicated by extensive neuroimaging studies (broadly considered as part of the pain matrix) [47]. Because certain brain structures scale with general head size, we employed estimated total intracranial volume (eTIV), scaling the volume of brain structures based on the individual’s intracranial volume [7]. We obtained the eTIV for each subject to investigate whether a group difference of regional [11C]Martinostat uptake was driven by ROI volumes relative to individual subjects.
PET image processing and analysis
For the full cohort, PET data in units of SUV spanning 60–90 mins after radiotracer administration (SUV60–90min) were used, while the full 0–90 mins data were used for subjects who have arterial blood data available. Preprocessing of the PET data included the following steps: Reconstructed PET images were first spatially smoothed by a Gaussian kernel of 6 mm full width at half maximum (FWHM). For the motion correction, we binned 30 minutes into 6 frames of 5 minutes and aligned them to a mean volume image using rigid body linear registration (6 d.o.f) using FSL MCFLIRT function (FMRIB (Oxford Centre for Functional MRI of the Brain) Software Library, Oxford, UK; https://fsl.fmrib.ox.ac.uk/fsl/) [26]. Using this linear transformation, unsmoothed PET data were aligned and averaged into a single frame. The frame-averaged PET image for each subject was then used to derive a linear transform to the T1-weighted structural scan using the boundary-based registration (bbregister) from FreeSurfer (version 6.0; http://surfer.nmr.mgh.harvard.edu)[13,19]. To account for partial volume effects in the PET images, a region-based, voxel-wise (RBV) method was applied with a geometric transfer matrix (GTM) derived from the segmentation of the T1-weighted image for each subject with tools implemented in PETSurfer. Lastly, the MEMPRAGE was registered to Montreal Neurological Institute (MNI152) atlas using linear (FLIRT (FMRIB’s linear image registration tool) and nonlinear (FNIRT (FMRIB’s nonlinear image registration tool) algorithms implemented in FSL [42]. Combining these transformation matrices, PET images were normalized to the MNI152 space and spatially smoothed by a Gaussian kernel of 8 mm (FWHM) for further voxel-based analyses. For region-based analyses, regional SUV or radioactivity values were extracted from the final unsmoothed, PVC-corrected PET data using the set of subject-specific ROIs in the native PET/MRI space. To facilitate intersubject comparison of regional HDAC distribution, we normalized regional SUV60–90min to individual subjects’ pons SUV60–90min as normalized SUV60–90min ratio (SUVR60–90min, Pons), similar to our previous study in healthy volunteers, which showed less variability in test/retest scans [50].
Kinetic modeling analysis
Full kinetic analyses were performed to estimate the VT in the subset of subjects who underwent blood draws from the arterial line. Our previous study with healthy individuals reported that these image-based outcome measures are well correlated with the VT with low inter-subject variability [50]. However, the validity of SUV60–90min and SUVR60–90min,Pons have not been tested in cLBP cohort. Kinetic modeling was conducted using PMOD 3.9 (PMOD Technologies Ltd, Zurich, Switzerland), with 27 ROIs to derive VT. Time activity curves (TACs) for each subject were extracted using subject-specific ROIs derived from FreeSurfer (as described above). In our previous work, a two-tissue compartment model (2TCM) was shown as the best model to estimate VT (ml/cm3) for [11C]Martinostat [50]. In this work, we used the 2TCM with the metabolite-corrected arterial plasma as an input function, fitting the blood volume fraction and a delay term for the plasma input function. Parametric images were generated with the pixel-wise modeling tool (PXMOD) of PMOD 3.9. The radioactivity concentration images were registered to the MNI152 atlas space, spatially smoothed (8 mm Gaussian kernel), and analyzed with the metabolite-corrected arterial plasma using the voxel-based graphical analysis (Logan invasive; t* = 40 min), then the resulting VT parametric maps (n=5) were averaged.
Statistical analysis
For region-based analysis, statistical tests were performed using R software (Version 4.2.3) and GraphPad Prism (Prism9, GraphPad Software Inc., San Diego, California, USA). The statistical analysis was performed in stepwise levels: 1) We compared SUV60–90 min of potential pseudo-reference regions (whole brain, white matter, deep white matter, pons, and cerebellum) between cLBP patients and controls, respectively, using a one-way analysis of variance (ANOVA) test. The aim is to identify a brain region with no significant group differences. 2) We assessed the association of SUV60–90 min or SUVR60–90min, Pons from the target ROIs with demographic variables (age, sex, BMI, and injected dose of the radiotracer) across the entire cohort using a non-parametric multivariate analysis of variance (MANOVA) with 5000 permutations, identifying demographic variables linked to PET measures (SUV60–90 min or SUVR60–90min, Pons). 3) We then examined the main effects of group (cLBP vs. controls) on SUV60–90 min or SUVR60–90min, Pons in the target ROIs through MANOVA with 5000 permutations. Any demographic variables showing significant associations with SUV60–90min or SUVR60–90min, Pons were factored in as covariates of no-interest in both MANOVA and subsequent post hoc analysis. 4) Should the MANOVA indicate significant group effect on PET measures (p < 0.05), we proceeded with post hoc multivariate analyses to delve deeper into these findings. 5) In cLBP patients, the relationship of SUV60–90 min or SUVR60–90min, Pons in the target ROIs with covariates of interest (pain, HDAS depression, and anxiety, ODI, and SOAPP-R) was tested using non-parametric MANOVA with 5000 permutations. 6) As an exploratory analysis, we conducted a multivariate post hoc analysis of the correlation between PET measures (SUV60–90 min or SUVR60–90min, Pons) in selected core ROIs (PAG, NAc, somatosensory, dlPFC) and pain intensity. 7) Lastly, a Pearson correlation analysis was performed to compare VT and image-derived PET measures (SUV60–90 min and SUVR60–90min, Pons) for all ROIs to assess validity of mage-based outcome measurements as a appropriate surrogate for those estimated with the full kinetic modeling.
A voxel-wise comparison for [11C]Martinostat uptake (SUV60–90min and SUVR60–90min, Pons) between cLBP patients and controls was performed using FSL’s FEAT (FMRIB software library) with an unpaired t-test (a significance threshold of Z > 2.3 with the cluster-based multiple comparison correction pcluster < 0.05). Whole-brain voxel-wise analyses correlating SUV60–90 min and SUVR60–90min, Pons values with the pain intensity were performed using FSL’s FEAT (Z > 2.3, pcluster < 0.05), with BMI added to the model as regressors of no interest.
Results
Demographics and clinical characteristics
Demographic information and clinical characteristics of participants are provided in Table 1. There were no significant differences in age, sex, or body mass index (BMI) between groups. However, the [11C]Martinostat injected dose differed significantly between cLBP and controls (p < 0.005). To address the potential confounding effect of the injected dose on group comparison, we performed an additional analyses. These consisted of comparing cLBP patients who received a lower dose (~5 mCi) with those injected with a higher dose (~15 mCi) (Supplementary Materials), and comparing a subset of matched cohorts (8 cLBP vs. 5 controls) with comparable injected doses (Supplementary Materials). With these additional analyses, we reassured that the variance in injected dose has minimal impact on our overall results.
Table 1.
Demographic characteristics, administered radiotracer dose, and cognitive metrics of study participants (mean ± SD).
| Demographic or survey metric | cLBP (n=23) | Control (n=11) | p-value |
|---|---|---|---|
|
| |||
| Age (year) | 47.96 ± 15.17 | 46.91 ± 15.26 | 0.8390 |
| Sex (M/F) | 11/12 | 7/4 | 0.6193 |
| Body Mass Index | 26.68 ± 4.91 | 26.73 ± 5.92 | 0.9798 |
| Injected dose (mCi) | 10.72 ± 4.32 | 6.10 ± 2.47 | 0.0026* |
| Pain Intensity | 4.48 ± 1.73 | n/a | |
| HADS - Depression (out of 21) | 3.87 ± 3.76 | n/a | |
| HADS - Anxiety (out of 21) | 5.78 ± 4.19 | n/a | |
| ODI (out of 49) | 11.48 ± 6.1 | n/a | |
| SOAPP-R (out of 96) | 10.78 ± 9.19 | n/a | |
Abbreviation: cLBP = chronic lower back pain patients; SD = standard deviation; HADS = Hospital Anxiety Depression Scale; ODI = Oswestry Disability Index; SOAPP-R= Screener and Opioid Assessment for Patients with Pain, Revised.
p<0.005.
[11C]Martinostat uptake between cLBP patients and matched controls
PET images of [11C]Martinostat are shown in Figure 1 to illustrate the spatial distributions of HDAC availability of cLBP patients and controls. Analysis revealed no significant correlation of either SUV60–90min or SUVR60–90min, Pons with age (F = 0.3933, p = 0.5557; F = 0.5739, p = 0.4965), sex (F = 1.4233; p = 0.2322; F = 0.3173, p = 0.7159), or injected dose (F = 1.0458, p = 0.3167; F = 0.6683, p = 0.4471). However, a significant association was found between BMI and both PET measures (SUV60–90min, F = 4.9892, p = 0.0288; SUVR60–90min, Pons, F = 4.4578, p = 0.0260). Therefore, we included BMI as a covariate alongside group effects in subsequent analyses. MANOVA demonstrated a significantly lower SUV60–90min (F = 5.5079; p = 0.0252) in cLBP patients compared to healthy control. A post hoc multivariate analysis indicated the regions showing significantly lower SUV60–90min included the thalamus, caudate, putamen, pallidum, hippocampus, amygdala, OFC, ACC, PCC, primary motor cortex, somatosensory cortex, dlPFC, precuneus, inferior frontal gyrus, somatosensory association cortex, middle temporal, superior parietal gyri (Figure S2(a)). Similarly, we found a significantly lower SUVR60–90min, Pons, in cLBP patients relatie to controls (F = 3.9708, p = 0.0368), with affected regions including ACC, PCC, primary motor cortex, somatosensory cortex, dlPFC, precuneus, inferior frontal gyrus, somatosensory association cortex, middle temporal, superior parietal gyri (Figure 2). Notably, estimated total intracranial volume (eTIV) and eTIV-normalized regional brain volumes did not differ between groups (Figure S3), which confirms that the observed [11C]Martinostat uptake differences were not driven by brain volumes (F = 0.5176, p = 0.5131).
Figure 1.

Group average maps of [11C]Martinostat uptake in SUV. SUV at 60–90 min post radiotracer injection (SUV60–90min) for cLBP patients (n=23) and age- and sex-matched healthy controls (n=11) are shown in the MNI152 atlas space. Standard uptake value, SUV; chronic low back pain, cLBP.
Figure 2.

Regional SUVR60–90min,Pons comparison between cLBP patients and matched controls (n=11). SUV at 60–90 min post radiotracer injection normalized to the pons SUV (SUVR60–90min,Pons) was plotted from selected brain regions. The main group effects (cLBP vs. controls) in SUVR60–90min, Pons in the target region of interests (n=22) were tested by using non-parametric multivariate analysis of variance (MANOVA) with 5000 permutations with the BMI as a covariates. If MANOVA indicated a significant group effect (p < 0.05), we performed addtional post hoc multivariate analyses. The asterisk indicates statistical significance, *p < 0.05, **p < 0.01. Standard uptake value, SUV; chronic low back pain, cLBP; analysis of variance, ANOVA.
In voxel-wise group comparison of SUV60–90min (Z > 2.3, pcluster < 0.05), cLBP patients showed significantly lower regional [11C]Martinostat uptake in multiple clusters within the somatosensory cortex, temporal, frontal cortex, cerebellum, and amygdala when compared to controls (Figure S2(b)). Nevertheless, no significant differences were observed in SUVR60–90min, Pons between cLBP patients and controls (Z > 2.3, pcluster < 0.05) when cluster-based multiple comparison corrections were applied. Moreover, no regions showed increased uptake (SUV60–90min and SUVR60–90min, Pons) in cLBP patients relative to controls.
Kinetic analysis
Due to equipment malfunction, blood data from six out of the eleven subjects who underwent arterial blood sampling were unable to be properly For the remaining subjects, radioactivity in the arterial plasma peaked within 1 min after radiotracer injection and decreased rapidly thereafter (Figure S4(a)). The averaged parent fraction ratios from cLBP patients (n=5) were about 0.5 at 30 min after radiotracer administration and eventually reached 0.2 by the end of the scan (Figure S4(b)). Consistent with our previous study in healthy individuals, the two-tissue compartment model (2TCM) best described dynamic [11C]Martinostat PET data obtained from cLBP patients [50]. Figure S4(c) shows representative time activity curves (TACs) and compartment model fitting results of the putamen from a representative cLBP patient. The 2TCM-estimated volume of distribution (VT) values ranged from 4.7 to 38.6 ml/cm3 (mean ± SD = 16.9 ± 7.1 across analyzed ROIs) (Table 2). Logan graphical analysis estimated regional VT values, using metabolite-corrected arterial blood as an input function (t* of 40 mins), yielded regional VT values that were positively correlated with those derived with 2TCM (Pearson r = 0.81, two-tailed p < 0.0001) (Table 2 and Figure 3 (a)). The VT ratio of Logan graphical analysis to 2TCM was 1.01 ± 0.17 (mean ± SD across all ROIs), indicating comparable estimates by the two methods. As illustrated in Figure S5, whole brain VT parametric map was calculated from the subset of the cLBP patients (n=5). We found a significant positive correlation between VT values and SUV60–90min (Pearson r = 0.72, two-tailed p < 0.0001; Figure 3(b)) and with SUVR60–90min, Pons (Pearson r = 0.72, two-tailed p < 0.0001; Figure 3(c)) in the subset of the cLBP cohort, confirming that SUV60–90min and SUVR60–90min, Pons are appropriate image-based substitutes for VT in cLBP patients. Similar to the previous study in healthy controls, SUVR60–90min, Pons demonstrated lower inter-subject variability, with a coefficient of variation (CV) ranging from 6.02 to 12.82%, compared to blood data–derived VT values (CV of 34.23 to 65.38%) across brain regions.
Table 2.
The volume of distribution (VT) estimated by using the 2TCM and Logan invasive method in the brain of cLBP patients (n=5).
| Brain regions | 2TCM | Logan invasive | ||
|---|---|---|---|---|
|
|
|
|||
| VT [mL/cm3] | SD | VT [mL/cm3] | SD | |
|
| ||||
| Whole brain | 15.20 | 6.15 | 16.88 | 8.73 |
| White matter | 14.28 | 5.32 | 16.05 | 7.61 |
| Deep white matter | 9.03 | 3.09 | 10.38 | 4.60 |
| Pons | 14.32 | 6.04 | 14.05 | 6.20 |
| Cerebellum | 22.63 | 11.21 | 22.33 | 13.12 |
| Brain stem | 13.50 | 5.37 | 13.28 | 5.33 |
| Thalamus | 17.08 | 7.68 | 19.20 | 11.64 |
| Caudate | 18.98 | 12.41 | 17.92 | 12.40 |
| Putamen | 22.27 | 9.56 | 23.49 | 12.41 |
| Pallidum | 17.94 | 7.82 | 17.94 | 9.05 |
| Hippocampus | 15.14 | 6.00 | 14.52 | 6.59 |
| Amygdala | 16.72 | 6.72 | 12.21 | 7.74 |
| Nucleus Accumbens | 21.20 | 10.14 | 20.57 | 11.56 |
| Periaqueductal gray | 15.33 | 6.69 | 12.38 | 3.34 |
| Orbitofrontal cortex | 19.73 | 11.37 | 21.52 | 14.72 |
| Anterior Cingulate Cortex | 18.63 | 7.69 | 20.25 | 10.29 |
| Posterior Cingulate Cortex | 17.76 | 6.18 | 18.67 | 8.48 |
| Insula | 18.85 | 7.78 | 19.37 | 8.86 |
| Precentral | 15.66 | 5.79 | 16.99 | 8.03 |
| Postcentral | 15.51 | 5.83 | 16.96 | 8.20 |
| Paracentral | 16.87 | 7.52 | 17.14 | 8.06 |
| Rostralmiddlefrontal | 17.37 | 7.10 | 19.16 | 9.83 |
| Precuneus | 17.44 | 6.25 | 19.17 | 8.80 |
| Inferiorfrontal gyrus | 16.91 | 6.19 | 17.38 | 7.61 |
| Suparmarginal | 17.51 | 6.48 | 18.93 | 9.07 |
| Middletemporal | 18.55 | 7.89 | 19.52 | 10.58 |
| Superiorparietal | 15.13 | 5.32 | 16.99 | 8.00 |
|
| ||||
| Averaged across ROIs | 17.02 | 7.31 | 17.53 | 8.90 |
Abbreviation: 2TCM = Two tissue compartment model; cLBP = chronic lower back pain patients; SD = standard deviation.
Figure 3.

Scatter plots of (a) regional VT values estimated using the 2TCM and Logan plot (t*=40 mins) and (b) regional VT values from the 2TCM and SUV60–90min or (c) SUVR60–90min, Pons in the subset of cLBP patients (n=5). Correlation analyses are conducted using a Pearson correlation analysis. Gray lines indicate standard deviation. Volume of distribution, VT; two tissue compartment model, 2TCM; standard uptake value at 60 to 90 minutes after a radiotracer injection, SUV60–90min; pons brain normalized standard uptake value, SUVR60–90min,Pons; chronic low back pain, cLBP.
Post hoc analyses
Post hoc analyses were performed to assess the relationship between regional [11C]Martinostat uptake from a priori ROIs and patient-reported outcome measures. We included the BMI as a covariate due to its established correlation with SUV60–90min and SUVR60–90min, Pons. Neither SUV60–90min nor SUVR60–90min, Pons showed a significant association with any self-report outcomes. However, considering potential region-specific associations, we conducted a focus examination of PET measures from selected brain regions (NAc, PAG, somatosensory cortex, dlPFC) and their relationship with pain intensity with a post hoc multivariate analysis. Interestingly, a trend level of higher reported pain intensity corresponded with reduced SUVR60–90min, Pons, particularly within the PAG (p = 0.0423) (Figure S6).
Discussion
Chronic pain is a complex disorder that affects both physical and mental functioning that could compromise quality of life. Current therapeutic strategies remain inadequate. Building on previous preclinical studies that implicate histone modifications are involved in animal pain models [16,49], we employed [11C]Martinostat PET to measure and compare in vivo HDAC expression levels in the brain of cLBP patients with age- and sex-matched healthy controls. In addition, we investigated the association between various patient-reported outcomes and HDAC availability in cLBP. Our findings indicated a pronounced decrease in HDAC availability in the cLBP patients compared to controls, particularly within the ACC, PCC, primary motor cortex, somatosensory cortex, dlFPC, precuneus, inferior frontal gyrus, somatosensory association cortex, middle temporal and superior parietal gyri. These results supported the hypothesis that HDAC availability was altered in cLBP, consistent with preclinical animal studies [2,9,16,49], providing evidence of HDAC alterations in vivo in the brains of cLBP patients.
We found that [11C]Martinostat SUV60–90min are significantly lower in the brain of cLBP patients than controls globally. A majority of the target ROIs, except for 5 brain regions (brain stem, NAc, PAG, insula, paracentral lobule), demonstrated significant group differences. When assessing SUVR60–90min, Pons, which is expected to have lower inter-subject variability than SUV60–90min and VT, we could identify the affected brain regions corresponding to the pain matrix [47]. Specifically, the primary motor cortex, somatosensory cortex, dlPFC, precuneus, inferior frontal gyrus, somatosensory association cortex, middle temporal, and superior parietal gyri in cLBP patients exhibited lower SUVR60–90min, Pons compared to controls. The thalamus, somatosensory cortex, superior parietal gyrus, somatosensory association cortex, cingulate cortices, and OFC are traditionally associated with the sensory component of pain [30,40,47]. The basal ganglia have also been implicated in pain processing based on preclinical and clinical evidence [3]. To ensure our results were valid, we explored potential confounding factors, including anatomical intracranial brain volumes, injected dose, age, and sex between cLBP and controls and confirmed no significant group differences. In addition, we found no significant correlation between PET measures (SUV60–90min and SUVR60–90min, Pons) and any patient-reported outcomes, potentially due to our relatively small sample size (n=23 cLBP). This is supported by our exploratory post hoc analysis which showed a trend level negative correlation between pain intensity and SUVR60–90min, Pons in the PAG (p = 0.0423). The PAG is a brain region with a high density of mu-opioid receptors and is considered a key component in the transmission and regulation of pain within the descending pain modulatory system and the processing of pain signals at the supraspinal level. While the MANOVA did not show a statistically significant association between pain and SUVR60–90min, Pons across the target ROIs (F = 0.8607, p = 0.3743), this trend in the PAG and pain intensity warrants further validation in a larger cohort.
Previous research has demonstrated a downward trend in histone modifications (H3K4me3, H3K27Ac, and H3K4me1) within the PAG, lateral hypothalamus, and NAc after a 5-week period post-sciatic nerve injury in mice [6]. This trend, specifically lower H3K4me1 and H3K4me3 levels in the PAG, correlates with individual variations in pain intensity [6], hinting at the enduring nature of epigenetic modifications in limbic brain structures in response to nerve injury and their potential role in sustained pain modulation. Inflammation induced by a complete Freund’s adjuvant (CFA) injection has been shown to the marginally reduce class I HDACs expression in the lumbar dorsal spinal cord [2]. Concordantly, other animal models have demonstrated differences in HDAC expression in response to nerve injury; for example, increased HDAC1 expression in the nuclear fraction of the ipsilateral lumbar spinal dorsal horn in a rat model of spinal nerve ligation (SNL) model [9], and a reduction of HDAC2 in the spinal dorsal horn 7 days after a spared nerve injury [41]. These findings supported the therapeutic exploration of HDAC inhibitors [1,16,17,36], which have demonstrated analgesic efficacy in animal pain models [2,9,14]. Collectively, these preclinical studies showed that efforts to examine changes in HDAC levels (or histone markers) primarily focus on the spinal cords while the supraspinal regions remain largely unexplored; furthermore, no study has previously investigated HDAC alternations in the brain of cLBP patients. Our study expands the understanding of HDAC involvement in chronic pain and reinforces the potential for HDAC modulation as a therapeutic target in humans.
Previous studies on histone modifications related to pain have yielded inconsistent results, although the trend has generally been towards elevated HDAC levels and reduced histone acetylation in animal models. This seems to contradict our findings in cLBP patients. However. direct comparisons are confounded not only by species differences but also by the chronicity of pain in our human cohort, as opposed to the acute or sub-acute pain often induced in animal studies. Furthermore, our study focused on cerebral HDAC, in contrast to the spinal-centric emphasis of most animal studies. It’s worth noting that our findings of altered HDAC availability in patients with various neurological and psychiatric disorders do not necessarily aligned with the directionality of HDAC levels suggested by preclinical research [37,45]. Beyond global alterations in HDAC expression and histone acetylation, targeted analyses focusing on the acetylation patterns of gene-specific promoters of pain-related receptors, ion channels, and cytokines, could provide additional insights on the role of histone modification in pain [10,28,29]. For example, neuropathic pain symptoms have been correlated with decreased expression of μ-opioid receptors and Nav1.8 sodium channels in the dorsal root ganglion [46], coinciding with enhanced histone acetylation in neuron-restrictive silencer factor promoter regions [33]. In addition, enhanced acetylation in the promoters of the genes like chemokine CC motif receptor 2 (CXCR2) was observed under pain conditions [43]. Regrettably, our current approach using [11C]Martinostat PET does not provide mechanistic evidence supporting specific modulation of gene expression. Nevertheless the observed reduction in HDAC availability within pain-associated brain regions of cLBP patients present new empirical evidence supporting the involvement of HDACs in chronic pain in the human brain. Future in vitro experiments elucidating the functional consequences of HDAC modulations on specific gene promoters related to pain-modulated receptors, ion channels, and cytokines may better illustrate the involvement of histone acetylation in different kinds of pain conditions.
We have validated the use of SUV60–90min and SUV60–90min, Pons as reliable image-based outcome measures to compare HDAC density between groups. Despite the challenge of variable injected doses, our analyses confirmed this factor has minimal impact on our overall results. However, we acknowledge that due to a relatively small sample size, sex differences in HDAC expression were not explored despite there are known differences in HDAC availability and the prevalence of chronic pain between males and females.
In conclusion, our study reported significant alteration in HDAC availability, indicated by [11C]Martinostat PET, in several brain regions associated with pain processing in cLBP patients, underscoring the potential of HDAC modulation as a target for chronic pain management. Future research with larger cohorts or in other pain conditions are essential to further elucidate the therapeutic implications targeting HDACs.
Supplementary Material
Acknowledgements
We thank Grae Arabasz, Shirley Hsu, Regan Butterfield, and Oliver Ramsay for their assistance with radiotracer administration, and the Radiopharmacy at the Martinos Center for Biomedical Imaging for support in processing arterial blood samples. The study was, in part, supported by National Institutes of Health (NIH) grants UH2AR076741 and R61DA048485 to H.-Y.W. Funding for the imaging facilities and infrastructure used in this work was provided by NIH grants S10RR017208, S10RR026666, S10RR022976, S10RR019933, S10RR023401, and S10OD023517.
Footnotes
Conflict of Interest.
The authors declare no competing financial interests.
The data supporting the findings of this study are available upon reasonable request.
Reference
- [1].Abel T, Zukin RS. Epigenetic targets of HDAC inhibition in neurodegenerative and psychiatric disorders. Curr Opin Pharmacol 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of class II histone deacetylases in the spinal cord attenuates inflammatory hyperalgesia. Mol Pain 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Borsook D, Upadhyay J, Chudler EH, Becerra L. A key role of the basal ganglia in pain and analgesia - insights gained through human functional imaging. Mol Pain 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Boudreau D, Von Korff M, Rutter CM, Saunders K, Ray GT, Sullivan MD, Campbell CI, Merrill JO, Silverberg MJ, Banta-Green C, Weisner C. Trends in long-term opioid therapy for chronic non-cancer pain. Pharmacoepidemiol Drug Saf 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Bruehl S, Burns JW, Passik SD, Gupta R, Buvanendran A, Chont M, Schuster E, Orlowska D, France CR. The Contribution of Differential Opioid Responsiveness to Identification of Opioid Risk in Chronic Pain Patients. J Pain 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Bryant S, Balouek JA, Geiger LT, Barker DJ, Peña CJ. Neuropathic pain as a trigger for histone modifications in limbic circuitry. Genes, Brain Behav 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Buckner RL, Head D, Parker J, Fotenos AF, Marcus D, Morris JC, Snyder AZ. A unified approach for morphometric and functional data analysis in young, old, and demented adults using automated atlas-based head size normalization: Reliability and validation against manual measurement of total intracranial volume. Neuroimage 2004. [DOI] [PubMed] [Google Scholar]
- [8].Bushnell MC, Čeko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nat Rev Neurosci 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Cherng CH, Lee KC, Chien CC, Chou KY, Cheng YC, Hsin ST, Lee SO, Shen CH, Tsai RY, Wong CS. Baicalin ameliorates neuropathic pain by suppressing HDAC1 expression in the spinal cord of spinal nerve ligation rats. J Formos Med Assoc 2014. [DOI] [PubMed] [Google Scholar]
- [10].Chiechio S, Copani A, Zammataro M, Battaglia G, IV RWG, Nicoletti F. Transcriptional regulation of type-2 metabotropic glutamate receptors: An epigenetic path to novel treatments for chronic pain. Trends Pharmacol Sci 2010. [DOI] [PubMed] [Google Scholar]
- [11].Compton WM, Volkow ND. Major increases in opioid analgesic abuse in the United States: Concerns and strategies. Drug Alcohol Depend 2006. [DOI] [PubMed] [Google Scholar]
- [12].Dagenais S, Caro J, Haldeman S. A systematic review of low back pain cost of illness studies in the United States and internationally. Spine J 2008. [DOI] [PubMed] [Google Scholar]
- [13].Dale AM, Fischl B, Sereno MI. Cortical surface-based analysis: I. Segmentation and surface reconstruction. Neuroimage 1999. [DOI] [PubMed] [Google Scholar]
- [14].Denk F, Huang W, Sidders B, Bithell A, Crow M, Grist J, Sharma S, Ziemek D, Rice ASC, Buckley NJ, McMahon SB. HDAC inhibitors attenuate the development of hypersensitivity in models of neuropathic pain. Pain 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Denk F, McMahon SB. Chronic pain: Emerging evidence for the involvement of epigenetics. Neuron 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Descalzi G, Ikegami D, Ushijima T, Nestler EJ, Zachariou V, Narita M. Epigenetic mechanisms of chronic pain. Trends Neurosci 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Géranton SM, Tochiki KK. Could targeting epigenetic processes relieve chronic pain states? Curr Opin Support Palliat Care 2015. [DOI] [PubMed] [Google Scholar]
- [18].Gilbert TM, Zürcher NR, Catanese MC, Tseng CEJ, Di Biase MA, Lyall AE, Hightower BG, Parmar AJ, Bhanot A, Wu CJ, Hibert ML, Kim M, Mahmood U, Stufflebeam SM, Schroeder FA, Wang C, Roffman JL, Holt DJ, Greve DN, Pasternak O, Kubicki M, Wey HY, Hooker JM. Neuroepigenetic signatures of age and sex in the living human brain. Nat Commun 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Greve DN, Fischl B. Accurate and robust brain image alignment using boundary-based registration. Neuroimage 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Gudin J, Kaufman AG, Datta S. Are opioids needed to treat chronic low back pain? A review of treatment options and analgesics in development. J Pain Res 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O’Neal L, McLeod L, Delacqua G, Delacqua F, Kirby J, Duda SN. The REDCap consortium: Building an international community of software platform partners. J Biomed Inform 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. A metadata-driven methodology and workflow process for providing translational research informatics support, J Biomed Inform. J Biomed Inf 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Ichise M, Toyama H, Innis RB, Carson RE. Strategies to improve neuroreceptor parameter estimation by linear regression analysis. J Cereb Blood Flow Metab 2002. [DOI] [PubMed] [Google Scholar]
- [24].Izquierdo-Garcia D, Hansen AE, Förster S, Benoit D, Schachoff S, Fürst S, Chen KT, Chonde DB, Catana C. An SPM8-based approach for attenuation correction combining segmentation and nonrigid template formation: Application to simultaneous PET/MR Brain Imaging. J Nucl Med 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Jang JH, Song EM, Do YH, Ahn S, Oh JY, Hwang TY, Ryu Y, Jeon S, Song MY, Park HJ. Acupuncture alleviates chronic pain and comorbid conditions in a mouse model of neuropathic pain: the involvement of DNA methylation in the prefrontal cortex. Pain 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Jenkinson M, Bannister P, Brady M, Smith S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage 2002. [DOI] [PubMed] [Google Scholar]
- [27].Ji RR, Kohno T, Moore KA, Woolf CJ. Central sensitization and LTP: Do pain and memory share similar mechanisms? Trends Neurosci 2003. [DOI] [PubMed] [Google Scholar]
- [28].Kiguchi N, Kobayashi Y, Saika F, Kishioka S. Epigenetic upregulation of CCL2 and CCL3 via histone modifications in infiltrating macrophages after peripheral nerve injury. Cytokine 2013. [DOI] [PubMed] [Google Scholar]
- [29].Kuehn BM. Opioid prescriptions soar: Increase in legitimate use as well as abuse. J Am Med Assoc 2007. [DOI] [PubMed] [Google Scholar]
- [30].Liang M, Su Q, Mouraux A, Iannetti GD. Spatial patterns of brain activity preferentially reflecting transient pain and stimulus intensity. Cereb Cortex 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Manchikanti L, Vallejo R, Manchikanti IV KN, Benyamin RM, Datta S, Christo PJ. Effectiveness of long-term opioid therapy for chronic non-cancer pain. Pain Physician 2011. [PubMed] [Google Scholar]
- [32].Manek NJ, MacGregor AJ. Epidemiology of back disorders: Prevalence, risk factors, and prognosis. Curr Opin Rheumatol 2005. [DOI] [PubMed] [Google Scholar]
- [33].Matsushita Y, Araki K, Omotuyi OI, Mukae T, Ueda H. HDAC inhibitors restore C-fibre sensitivity in experimental neuropathic pain model. Br J Pharmacol 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Mauck MC, Lotz J, Psioda MA, Carey TS, Clauw DJ, Majumdar S, Marras WS, Vo N, Aylward A, Hoffmeyer A, Zheng P, Ivanova A, McCumber M, Carson C, Anstrom KJ, Bowden AE, Dalton D, Derr L, Dufour J, Fields AJ, Fritz J, Hassett AL, Harte SE, Hue TF, Krug R, Loggia ML, Mageswaran P, McLean SA, Mitchell UH, O’Neill C, Pedoia V, Quirk DA, Rhon DI, Rieke V, Shah L, Sowa G, Spiegel B, Wasan AD, Wey(Monica) H-Y, LaVange L. The Back Pain Consortium (BACPAC) Research Program: Structure, Research Priorities, and Methods. Pain Med 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Mehta SP, Crochet Z, O’Dell N, Stevenson A. Hospital anxiety and depression scale. Crit Rev Phys Rehabil Med 2018. [Google Scholar]
- [36].Micelli C, Rastelli G. Histone deacetylases: Structural determinants of inhibitor selectivity. Drug Discov Today 2015. [DOI] [PubMed] [Google Scholar]
- [37].Pascoal TA, Chamoun M, Lax E, Wey HY, Shin M, Ng KP, Kang MS, Mathotaarachchi S, Benedet AL, Therriault J, Lussier FZ, Schroeder FA, DuBois JM, Hightower BG, Gilbert TM, Zürcher NR, Wang C, Hopewell R, Chakravarty M, Savard M, Thomas E, Mohaddes S, Farzin S, Salaciak A, Tullo S, Cuello AC, Soucy JP, Massarweh G, Hwang H, Kobayashi E, Hyman BT, Dickerson BC, Guiot MC, Szyf M, Gauthier S, Hooker JM, Rosa-Neto P. [11C]Martinostat PET analysis reveals reduced HDAC I availability in Alzheimer’s disease. Nat Commun 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Ploghaus A, Narain C, Beckmann CF, Clare S, Bantick S, Wise R, Matthews PM, Nicholas P Rawlins J, Tracey I. Exacerbation of pain by anxiety is associated with activity in a hippocampal network. J Neurosci 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Schroeder FA, Wang C, Van De Bittner GC, Neelamegam R, Takakura WR, Karunakaran A, Wey HY, Reis SA, Gale J, Zhang YL, Holson EB, Haggarty SJ, Hooker JM. PET imaging demonstrates histone deacetylase target engagement and clarifies brain penetrance of known and novel small molecule inhibitors in rat. ACS Chem Neurosci 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Senba E, Kami K. A new aspect of chronic pain as a lifestyle-related disease. Neurobiol Pain 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Shen X, Liu Y, Xu S, Zhao Q, Wu H, Guo X, Shen R, Wang F. Menin regulates spinal glutamate-GABA balance through GAD65 contributing to neuropathic pain. Pharmacol Reports 2014. [DOI] [PubMed] [Google Scholar]
- [42].Smith SM, Jenkinson M, Woolrich MW, Beckmann CF, Behrens TEJ, Johansen-Berg H, Bannister PR, De Luca M, Drobnjak I, Flitney DE, Niazy RK, Saunders J, Vickers J, Zhang Y, De Stefano N, Brady JM, Matthews PM. Advances in functional and structural MR image analysis and implementation as FSL. NeuroImage.2004. [DOI] [PubMed] [Google Scholar]
- [43].Sun Y, Sahbaie P, Liang DY, Li WW, Li XQ, Shi XY, Clark JD. Epigenetic regulation of spinal cxcr2 signaling in incisional hypersensitivity in mice. Anesthesiology 2013. [DOI] [PubMed] [Google Scholar]
- [44].Tochiki KK, Cunningham J, Hunt SP, Géranton SM. The expression of spinal methyl-CpG-binding protein 2, DNA methyltransferases and histone deacetylases is modulated in persistent pain states. Mol Pain 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Tseng CEJ, Gilbert TM, Catanese MC, Hightower BG, Peters AT, Parmar AJ, Kim M, Wang C, Roffman JL, Brown HE, Perlis RH, Zürcher NR, Hooker JM. In vivo human brain expression of histone deacetylases in bipolar disorder. Transl Psychiatry 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies C-Fiber dysfunctions in neuropathic pain. J Neurosci 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Wager TD, Atlas LY, Lindquist MA, Roy M, Woo C-W, Kross E. An fMRI-Based Neurologic Signature of Physical Pain. N Engl J Med 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Wang C, Schroeder FA, Wey HY, Borra R, Wagner FF, Reis S, Kim SW, Holson EB, Haggarty SJ, Hooker JM. In vivo imaging of histone deacetylases (HDACs) in the central nervous system and major peripheral organs. J Med Chem 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Wang W, Cui S shan, Lu R, Zhang H. Is there any therapeutic value for the use of histone deacetylase inhibitors for chronic pain? Brain Res Bull 2016. [DOI] [PubMed] [Google Scholar]
- [50].Wey HY, Gilbert TM, Zürcher NR, She A, Bhanot A, Taillon BD, Schroeder FA, Wang C, Haggarty SJ, Hooker JM. Insights into neuroepigenetics through human histone deacetylase PET imaging. Sci Transl Med 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Wey HY, Wang C, Schroeder FA, Logan J, Price JC, Hooker JM. Kinetic Analysis and Quantification of [11C]Martinostat for in Vivo HDAC Imaging of the Brain. ACS Chem Neurosci 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Woolf CJ, Mannion RJ. Neuropathic pain: Aetiology, symptoms, mechanisms, and management. Lancet 1999. [DOI] [PubMed] [Google Scholar]
- [53].Zgierska AE, Ircink J, Burzinski CA, Mundt MP. Cost of opioid-treated chronic low back pain: Findings from a pilot randomized controlled trial of mindfulness meditation-based intervention. J Opioid Manag 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Zhuo M Cortical excitation and chronic pain. Trends Neurosci 2008. [DOI] [PubMed] [Google Scholar]
- [55].Ziv M, Tomer R, Defrin R, Hendler T. Individual sensitivity to pain expectancy is related to differential activation of the hippocampus and amygdala. Hum Brain Mapp 2010. [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.
