Текст
                    iew
ed

White matter microstructure and serum biomarkers of inflammation in functional seizures

Christina Mueller, PhD & Jerzy P. Szaflarski, MD, PhD

Address for correspondence:

ev

CIRC 312, 1719 6th Ave S, Birmingham, AL 35233

er
r

P: 205-975-4219 | Fax: 205-996-4802 | e-mail: cm1@uab.edu or jszaflarski@uabmc.edu

Author affiliation:

University of Alabama at Birmingham (UAB) Heersink School of Medicine, Department of

Pr

ep

rin

tn

ot

pe

Neurology and the UAB Epilepsy Center, Birmingham, AL, USA

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791


iew ed Abstract Background: Neuroinflammation may contribute to the pathophysiology of functional seizures (FS). However, it is unclear whether and to what degree comorbid psychiatric symptoms explain this association. In this study, we investigated the neuroinflammatory signature specific to FS ev and how it compared to that of psychiatric controls (PCs). Methods: We prospectively assessed differences in neurite density (NDI), orientation dispersion er r (ODI), and isotropic diffusion (F-ISO) between 23 participants with FS and 27 PCs, and their relationships to serum levels of tumor necrosis factor (TNF)-, TNF receptor 1 (TNF-R1), TNFrelated apoptosis-inducing ligand (TRAIL), interleukin (IL)-6, intercellular adhesion molecule pe (ICAM)-1, and monocyte chemoattractant protein (MCP)-1 using voxelwise multiple linear regressions. Pearson correlations between serum biomarkers and clinical symptoms were also obtained. ot Results: There were no WM microstructural differences between groups. In FS, TNF-R1 was negatively associated with NDI in the right UF and positively associated with F-ISO in the left tn uncinate fasciculus (UF). IL-6 was positively associated with NDI and negatively with F-ISO in the left UF. ICAM-1 was positively associated with ODI in the left UF. TNF- was negatively rin associated with ODI in the left cingulum bundle. The opposite relationships were observed in PCs. Higher TNF-R1 was associated with higher depression, anxiety, lower emotional quality of ep life, and higher levels of disability in FS. Conclusions: For the first time, we report relationships between peripheral inflammatory Pr biomarkers and WM integrity in FS including abnormalities in the UF and cingulum bundle. Our results suggest that serum biomarkers of inflammation may, with additional studies, become a This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed useful aid to FS diagnosis, especially in settings where video-EEG is not available. The lack of group differences in WM microstructure suggests that previously identified WM abnormalities in FS versus healthy controls may be related to psychological comorbidities of FS. Key words: functional seizures, neuroinflammation, neurite orientation dispersion and density Pr ep rin tn ot pe er r ev imaging, white matter, psychiatric comorbidities This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Introduction Functional seizures (FS) are a subtype of functional neurological symptom disorder (FNSD) involving seizure-like episodes without the presence of epileptiform discharges. Similar to other neuropsychiatric conditions such as depression, anxiety, and posttraumatic stress ev disorder (PTSD), the pathophysiology of FS may involve abnormal immune system functioning and neuroinflammation (NI). 1 In the recently proposed two-hit hypothesis of FS, we suggested er r early life stress (physical or psychological), structural abnormalities, and brain inflammation as potential factors in FS development and maintanance. 1 Within this model, trauma induces neuroendocrine (e.g., hypothalamic-pituitary axis activation (HPAA), cortisol release) and pe neuroinflammatory (e.g., immune cell activation and pro-inflammatory cytokine release) changes that prime the brain toward NI in response to subsequent traumatic events. Gledhill et al. 2 recently assessed inflammatory markers in FS, epilepsy, and healthy ot controls (HCs) to report elevated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and intercellular adhesion molecule (ICAM)-1 following FS compared to epileptic tn seizures, lending support to the two-hit hypothesis of FS. In further support of the chronic HPAA model, a recent meta-analysis in FS found elevated baseline heart rate, peri-ictal cortisol, and rin adrenocorticotropic hormone, and decreased brain-derived neurotropic factor (BDNF). 3 In response to systemic inflammation, the liver secretes C-reactive protein, 4 which is used ep clinically as a marker of inflammation. One study reported C-reactive protein elevations in adolescents with FS compared to HCs, with levels being similar to other FNSD types (motor, Pr sensory, and mixed). 5 Brain micro-pathology is common in systemic inflammatory disorders, 6-9 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
inflammation and white matter (WM) structure in FS. 10 iew ed but studies to date have not assessed the relationship between peripheral biomarkers of Neurite orientation dispersion and density imaging (NODDI) is a multi-shell diffusion- weighted imaging approach that quantifies neurite density (neurite density index, NDI), axonal coherence (orientation dispersion index, ODI), and the fraction of isotropic (free water) diffusion ev (F-ISO) in the brain. 11 The NDI has been histologically validated to reflect the density of axons and dendrites in the brain, and their loss in disease. 12,13 The ODI has been similarly validated on er r histology, with lower ODI indicating damaged white matter (WM). 13 NODDI is an extension of conventional diffusion imaging approaches such as diffusion tensor imaging (DTI), which have shown diminished WM integrity in sensorimotor, attention, and emotion regulation networks in pe FS, including in the uncinate fasciculus (UF), superior temporal gyrus, and the insula. 14-17 One study from our group has applied NODDI to a large sample of FS patients to report decreased NDI in the cingulum bundle, UF, fornix/stria terminalis (FST), and corticospinal tract (CST) in ot FS compared to controls. 18 One of the most prominent criticisms of currently available FS studies is that tn comparisons are typically made to HCs rather than to participants with similar psychiatric comorbidities or to organic counterpart conditions (e.g., epileptic seizures) to facilitate more in- rin depth evaluation of the underlying neuropathology. 19 First attempts to address this shortcoming have been made recently by controlling for history of TBI and by comparing FS to epilepsy. 18,20 ep However, it remains unclear whether the observed WM abnormalities are specific to FS or related to psychiatric symptoms in general. NI is well documented in psychiatric disorders Pr including depression, anxiety, and PTSD. 21,22 Due to the high prevalence of these disorders in FS, a neuroinflammatory signature specific to FS needs to be defined. 3,23 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Thus, the first aim of the current study was to assess whether FS involves WM microstructural abnormalities that do not exist in other mental health disorders (MHDs). Based on the results from the literature, if such differences exist, we expected to observe decreased NDI and ODI and increased FW in the cingulum bundle, UF, FST, and CST in FS when compared to a group of patients with depression, anxiety, and PTSD, confirming that previously identified ev WM abnormalities are specific to FS rather than psychopathology in general. 19,24 Secondly, we wanted to assess the relationship between peripheral inflammatory markers and WM er r microstructure in the two groups with a specific focus on the same WM tracts. 11,18 Because axonal loss and edema can result from NI, we expected to observe significant relationships between the NDI and F-ISO and the serum biomarkers. 25-27 Lastly, we aimed to assess whether pe inflammatory biomarkers are predictive of FS symptoms, hypothesizing that increased levels of all biomarkers would be associated with more severe symptoms. 21,28-32 To achieve these aims, we first compared NODDI metrics in 23 patients with FS and 27 ot psychiatric controls (PCs) and then assessed associations between NODDI indices and serum levels of tumor necrosis factor (TNF)-, TNF receptor 1 (TNF-R1), TNF-related apoptosis- tn inducing ligand (TRAIL), interleukin (IL)-6, intercellular adhesion molecule (ICAM)-1, and monocyte chemoattractant protein (MCP)-1. Finally, we assessed bivariate correlations between rin the serum biomarkers and clinical symptoms. TRAIL and ICAM-1 were chosen because they were previously found to distinguish FS ep from epileptic seizures. 2 Due to their association with FS, we expected both to be higher in the FS group compared to PCs. TNF- is a pro-inflammatory cytokine that promotes cell Pr proliferation that was found to be elevated in psychiatric disorders involving inflammation, and would likely be higher in PCs compared to FS. 32-35 TNF-R1 is a TNF- receptor that is similarly This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed elevated in inflammatory psychiatric disorders, particularly bipolar disorder, 36 and likely higher in PCs than FS. IL-6 is a pro-inflammatory cytokine capable of crossing the blood-brain-barrier that is potentially elevated in PCs compared to FS. 30 Lastly, MCP-1 is a chemokine that regulates monocyte and macrophage migration and infiltration into the brain28,37 and is potentially involved in the pathophysiology of depression. 29 We expected MCP-1 to be elevated er r Methods ev in PCs compared to FS. Design The current study was a prospective, cross-sectional observational study examining WM pe microstructure and serum inflammatory biomarkers in FS and PCs. ot Participants We recruited participants with FS between the ages of 15 and 60 from an outpatient tn psychology clinic and from the Epilepsy Monitoring Unit at the University of Alabama at Birmingham (UAB). A definite diagnosis of FS38 confirmed through video-EEG without rin comorbid epilepsy was required for participation. Age- and sex-matched PCs with MHDs were also prospectively recruited through an online advertisement on the UAB clinical trials website. Participants underwent phone screening to confirm inclusion and exclusion criteria, including ep medical history, current medications, and MRI contraindications. A partial waiver of consent was obtained for the phone screening. Pr The following criteria were exclusionary in both groups: 1. autoimmune, inflammatory, or neurological conditions (e.g., epilepsy, stroke, multiple sclerosis), 2. pregnancy or lactation, 3. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed MRI contraindications, 4. psychotic illness (e.g., schizophrenia, schizophreniform disorder, brief psychotic disorder), 5. personality disorder, 6. substance use disorder, and 7. inability to complete study procedures. Study Protocol ev The study received approval from the UAB Institutional Review Board and written informed consent was obtained from participants prior to study procedures. Children under 18 er r provided assent, and parents or legal guardians provided consent. The work was carried out in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki). Pregnancy testing was performed in women of reproductive age. pe The following questionnaires were administered: demographics questionnaire, Hospital Anxiety and Depression Scale (HADS), 39 Dissociative Experience Scale, 2nd edition (DES-II), 40 Brief Trauma Questionnaire (BTQ), 41 Holmes-Rahe Life Stress Inventory/Social Readjustment ot Rating Scale, 42 Symptom Checklist 90 (SCL-90), 43 Somatoform Dissociation Questionnaire (SDQ-20), 44 Quality of Life in Epilepsy (QOLIE-31), 45 and the WHO Disability Assessment tn Schedule version 2.0 (WHODAS 2.0). 46 Participants with FS also reported FS over the previous seven days using a semi-structured interview. rin Ten milliliters of blood were drawn from the antecubital fossa into serum separation tubes. Serum was separated by centrifugation and stored at -80C at the UAB Center for Clinical ep and Translational Sciences. Serum levels of TNF-R1 and TRAIL were determined with R&D ELISA kits (Minneapolis, MN) in the UAB Metabolism Core Laboratory. IL-1, IL-6, TNF-, Pr ICAM)-1, and MCP-1 were quantified with multiplex electrochemiluminescence using a Meso Scale Discovery QuickPlex SQ 120 imager (MSD; Rockville, MD). This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Neuroimage Acquisition Participants completed neuroimaging on a 3-Tesla Siemens Prisma Magnetom with 20channel head coil (Siemens Healthineers, Erlangen, Germany). We collected two anatomical sequences. The first one was a T1-weighted high-resolution Magnetization Prepared Rapid ev Gradient Echo (MPRAGE) scan acquired with TR 2.4s, TE 2.22ms, 8 flip angle, 0.8mm slice thickness, 208 slices, 256×256 matrix, 0.8mm3 voxel resolution. The second was a T2-weighted er r scan acquired with TR 3.2s, TE 563ms, GRAPPA acceleration factor 2, 0.8 mm slice thickness, 192 slices, 256×240mm field-of-view, 320×300 matrix, and 0.8 mm3 voxel resolution. Diffusion scans were two identical multi-shell diffusion-weighted sequences (b-values centered around pe 1500s/mm2 and 3000 s/mm2). These were acquired in opposite (anterior-posterior and posterioranterior) phase encoding directions with TR 3230ms, TE 89.2ms, multi-band acceleration factor 4, 78 flip angle, 160 refocus flip angle, 1.5mm slice thickness, 92 slices, 210×210mm field-of- ot view, 140×140 matrix, 1.5mm3 voxel resolution, 92 repetitions (46 with each of the two b- repetitions. tn values). Seven b0 images were also acquired, which were interleaved with the diffusion-weighted rin Neuroimage Processing Preprocessing of structural and dMRI data was performed with AFNI version 20.1.18. The anatomical images were skull-stripped (3dSkullStrip), deobliqued (3dWarp -deoblique), ep 47,48 and axialized (fat_proc_axialize_anat). Next, the dMRI images were co-registered with the Pr axialized T2-weighted image (3dAllineate) and then processed with the DIFFPREP module of the NIH TORTOISE toolbox, version 3.2, 49,50 which consisted of motion correction and eddy This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed current removal. Blip-up/blip-down (reversed phase encoding) EPI distortion correction was performed with the DR-BUDDI module of TORTOISE, 51 which uses geometric averaging to combine the two dMRI sequences acquired in opposite phase-encoding directions. A whole-brain mask was created using the T2-weighted image (3dAutomask) and resampled to match the resolution of the dMRI data (3dresample). Negative values were ev removed from the dMRI images, and the images were normalized by dividing them by the first b0 image in the sequence. The NODDI three-compartment model was fitted to the dMRI data er r using the NODDI toolbox, version 1.05 (available from nitrc.org) for Matlab, version 2020a (Mathworks, Natick, MA), resulting in whole-brain NDI, ODI, and F-ISO maps. The NDI maps were thresholded at 0.95 to reduce the impact of EPI distortion artifacts at the frontal, temporal, pe and occipital poles. Both the DTI and NODDI maps were smoothed (AFNI’s 3dMedianFilter with 3mm Gaussian kernel) and transformed into Montreal Neurological Institute (MNI) 2mm space by first warping the T1-weighted image to standard space (3dQwarp) and then applying tn Statistical Analyses ot the warp to the diffusion maps (3dNwarpApply). Group differences in demographic information, questionnaire data, and serum biomarkers rin were tested with independent-samples t-tests (continuous variables) and Chi-Square tests (categorical variables) in IBM SPSS Statistics for Mac, Version 28.0 (IBM Corp, Armonk, NY). ep Statistical significance was assumed at p<0.05 (two-tailed). Voxelwise group differences in NODDI metrics were assessed with Analyses of Covariance (ANCOVAs) within AFNI’s Pr 3dttest++, with all six serum biomarker levels (IL-6, TNF-, ICAM-1, MCP-1, TNF-R1, and TRAIL) added as covariates of interest. Because all serum biomarkers were added to the regression model This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed simultaneously, the reported associations are the unique variance contributed by each biomarker. Three participants had undetectable levels (<0.25 pg/ml) of IL-6 and those values were set to zero for the regressions. No other metabolite levels were undetectable or missing. A mask was applied to restrict the analyses to the cerebral WM. Cluster thresholds for each outcome were determined by performing 3dFWHMx on the residuals from the models, which estimates the ev spatial noise of the error. The estimate was used with 3dClustSim to determine the following cluster thresholds for nearest-neighbor 1 clustering and bi-sided thresholding with a voxel-level er r threshold of p=0.01 and cluster threshold of =0.05: NDI >657, ODI >664, and F-ISO >844 voxels. Bi-sided thresholding ensured that adjacent clusters with positive (FS>PCs) and negative (FS<PCs) group differences were reported as separated clusters. Cluster thresholds reduce the detect areas of significance. pe number of false positives in mass univariate (voxelwise) analyses while retaining sensitivity to For visualization of interaction effects, mean values in significant clusters were extracted ot from each participant’s image with AFNI’s 3dROIstats function, and plotted against inflammatory biomarker levels. tn Lastly, Pearson correlation coefficients were obtained between serum biomarkers and clinical outcomes. The analyses were corrected for the number of questionnaire scores (29) using rin a false discovery rate of 0.05, yielding a corrected p-value of 0.0126. 52 In the FS group, associations between serum biomarkers and seven-day FS count were assessed with Spearman Pr ep correlations at p<0.05, which are more appropriate for ordinal data. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Participant characteristics iew ed Results Eighty-four individuals with FS were screened and 42 met inclusion criteria. Thirteen withdrew prior to attending the study visit, five did not tolerate the MRI, and one did not complete the diffusion imaging protocol. Twenty-three FS participants provided usable data. ev Forty-seven PCs were screened, 31 were eligible, four withdrew prior to the study visit, and 27 provided usable data. Twenty-two FS patients and 25 PCs also provided blood and were included er r in the correlation analyses. Demographic information and questionnaire data are shown in Table 1. The groups did not differ in demographic variables. FS patients exhibited higher depressive symptoms on the pe HADS, worse quality of life (QoL) and higher levels of psychopathology in several domains, including somatization, more prevalent dissociative experiences, and more severe somatoform dissociation than PCs. The FS group endorsed higher levels of disability in all domains, ot including lower cognitive functioning and higher total disability. A higher proportion of FS participants compared to PCs met DSM-5 criteria for lifetime exposure to a traumatic event. All tn other outcomes, including stressful life events and anxiety, were not different between groups. Serum levels of IL-6, TNF-, ICAM-1, MCP-1, and TRAIL were not different between rin groups (p>0.05). TNF-R1 levels were higher in the FS group (mean=1359.95 pg/ml) compared Pr ep to PCs (mean=1202.94 pg/ml; t(45)=-2.155, p=0.037). This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
tn rin ep Pr 23 4 1 - 0 5 - 8 16 1 2.04 2.20 17 2 - p 0.668 0.507 1.747 0.627 - er r 4.57 4.65 4.74 10.11 3.48 3.92 3.127 1.380 0.003** 0.174 215.17 141.35 194.37 140.03 0.521 0.605 32.66 58.75 45.64 33.41 38.93 64.65 51.20 31.01 16.25 26.09 25.14 29.39 37.03 23.03 -68.89 63.11 44.81 69.37 94.15 89.81 -12.83 15.35 18.27 21.92 18.28 17.76 -2.441 0.018* † -2.775 0.009** -1.838 0.072 † -4.031 <0.001*** -3.301† 0.002** -6.627 <0.001*** 1.67 1.84 1.27 1.79 1.64 0.94 1.27 1.04 1.03 1.47 1.11 1.16 1.67 1.31 1.19 1.17 1.29 1.16 1.06 1.04 0.46 0.99 0.70 0.93 0.61 0.43 0.37 0.69 0.33 0.66 0.31 0.77 0.53 0.74 0.44 0.48 0.70 0.93 0.42 0.43 5.069† <0.001*** 2.991† 0.005** † 2.147 0.040* † 2.792 0.009** † 3.903 <0.001*** 1.979† 0.058 † 2.989 0.005** 1.188 0.241 † 2.959 0.006** 3.479† 0.002** 30.19 22.31 14.15 11.73 33.83 10.33 22.37 2.92 5.146† <0.001*** 36.36 38.07 22.58 25.87 17.41 8.10 13.33 13.52 3.475† 0.001** † 4.914 <0.001*** pe 8.30 11.78 t/X2 -0.431 0.440 ev - iew ed PC (N=27) Mean SD 38.67 12.78 21 2 ot Age Sex Female (N) Male (N) Race Asian (N) Black/African American (N) White (N) Other (N) Weekly FS count Questionnaires HADS Depression Anxiety LSI Total score QOLIE-31 Seizure Worry Overall QoL Emotional Energy Cognitive Medication Social SCL-90 Somatization Obsessive-Compulsive Interpersonal Sensitivity Depression Anxiety Anger-Hostility Phobic-Anxiety Paranoid Ideation Psychoticism Global Scale DES-II Total Score SDQ Total Score WHODAS 2.0 Cognition Mobility FS (N=23) Mean SD 37.17 11.48 3.047† 0.005** This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
er r ev iew ed Self-care 18.18 20.62 3.33 10.00 3.094† 0.004** † Getting Along 35.98 31.75 16.98 20.35 2.431 0.020* Life Activities 47.27 33.21 24.44 21.90 2.776† 0.009** Participation 49.62 27.12 18.36 22.92 4.373 <0.001*** Total Score 39.28 22.16 15.22 13.54 4.459† <0.001*** BTQ 5.571 0.018* Trauma Exposure (N) 22 21 No Trauma Exposure (N) 0 6 Not Reported (N) 1 0 Serum Biomarker IL-6 (pg/ml) 1.37 0.93 1.11 0.74 -1.047 0.301 1.61 0.39 1.56 0.52 -0.406 0.687 TNF- (pg/ml) † ICAM-1 (ng/ml) 590.23 184.47 520.73 102.99 -1.566 0.127 MCP-1 (pg/ml) 250.55 148.14 234.88 55.73 -0.468† 0.644 TNF-R1 (pg/ml) 1359.95 244.21 1202.94 253.47 -2.155 0.037* TRAIL (pg/ml) 96.75 20.43 95.91 27.06 -0.119 0.904 †df adjusted for unequal variances. ††higher scores reflect better quality of life. *p<0.05 **p<0.01 ***p<0.001 pe Table 1. Demographic information and questionnaire data for the final sample. BTQ=Brief Trauma Questionnaire, DES-II=Dissociative Experiences Scale, 2nd edition, HADS=Hospital Anxiety and Depression Scale, ICAM-1=intercellular adhesion molecule 1, IL=interleukin, ot LSI=Life Stress Inventory, MCP=monocyte chemoattractant protein, QoL=Quality of Life, QOLIE-31=Quality of Life in Epilepsy, 31-item version, SDQ-20=Somatoform Dissociation tn Questionnaire, TNF=tumor necrosis factor, TNF-R1=TNF receptor 1, TRAIL= TNF-related rin apoptosis-inducing ligand, WHODAS 2.0=WHO Disability Assessment Schedule, 2nd edition Main imaging results ep The ANCOVAs revealed no significant clusters where NDI, ODI, or F-ISO differed between groups. However, there were several significant cluster-level interactions between Pr serum inflammatory biomarkers and group on the NODDI outcomes (Table 2). This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Cluster size (mm3) UF Ant. corona radiata Ant. thalamic radiation Inf. fronto-occipital fasciculus L +11 2111 Ant. limb of internal capsule Ant. thalamic radiation Region cluster 2 cluster 3 cluster 4 cluster 5 -2 +2 870 L -24 +26 +11 7557 R +25 +32 +10 3462 R +18 +30 +44 2102 L -32 -13 +39 1744 L -17 +12 +54 1084 Ant. thalamic radiation Inf. fronto-occipital fasciculus ODI R +44 +41 -10 1349 Post. thalamic radiation Forceps major Inf. fronto-occipital fasciculus Inf. longitudinal fasciculus Unspecified WM in sup. parietal and lateral occipital lobe Cingulum bundle Body of corpus callosum L -31 -73 +7 1243 L -12 -40 +61 1092 L -9 -18 +36 705 er r UF Genu of corpus callosum Ant. limb of internal capsule Ant. corona radiata Ant. thalamic radiation Forceps Minor Inf. fronto-occipital fasciculus Genu of corpus callosum Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Unclassified WM adjacent to superior frontal gyrus Sup. corona radiata Sup. longitudinal fasciculus Unclassified WM adjacent to superior frontal gyrus rin group x TRAIL cluster 1 ep group x TNF- cluster 1 Pr cluster 2 cluster 3 +27 -21 pe group x TNFR1 cluster 1 -26 L ot group x MCP-1 cluster 1 tn group x IL-6 cluster 1 ev NDI iew ed Predictor MNI coordinates (peak signal) Hem. x y z This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
cluster 2 UF Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Post. thalamic radiation Forceps major Inf. fronto-occipital fasciculus Inf. longitudinal fasciculus 674 R +12 -64 +50 666 L -25 +32 +6 1080 -73 +4 680 Inf. fronto-occipital fasciculus F-ISO R +34 +51 -5 1604 Unclassified WM adjacent to inf. fronto-occipital fasciculus UF Ant. corona radiata Ant. thalamic radiation Inf. fronto-occipital fasciculus R +29 +48 -10 2670 L -27 +26 +10 1684 L -27 +28 +12 7206 R +25 +32 +8 3427 L -32 -13 +39 1823 pe ot UF Genu of corpus callosum Ant. internal capsule Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus External capsule Genu of corpus callosum Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Sup. corona radiata Sup. longitudinal fasciculus ep rin group x TNFR1 cluster 1 +16 -26 tn cluster 2 -25 L group x TRAIL group x IL-6 cluster 1 iew ed group x ICAM1 cluster 1 +30 ev cluster 5 R er r cluster 4 Splenium of corpus callosum Post. limb of internal capsule Retrolenticular part of internal capsule Post. corona radiata Post. thalamic radiation External capsule Unspecified WM in sup. parietal and lateral occipital lobe Pr cluster 2 cluster 3 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Unclassified WM adjacent to sup. frontal gyrus group x TRAIL cluster 1 R +13 +22 +55 Unclassified WM adjacent to frontal R +22 +61 -14 pole Table 2. Significant clusters with interactions between group and serum biomarkers. Note: Only tracts with cluster overlap of >3% are reported. 897 iew ed cluster 4 1836 Ant.=anterior, F-ISO=fraction of isotropic diffusion, Hem.=hemisphere, ICAM-1=intercellular ev adhesion molecule 1, IL-6=interleukin 6, Inf.=inferior, L=left, MCP-1=monocyte chemoattractant protein 1, NDI=neurite density index, MNI=Montreal Neurological Institute, er r ODI=orientation dispersion index, Post.=posterior, R=right, Sup.=superior, TNF-=tumor necrosis factor , TNF-R1=tumor necrosis factor receptor 1, TRAIL=tumor necrosis factor- ot Serum-by-group interactions pe related apoptosis-inducing ligand, WM=white matter IL-6 was positively associated with NDI in the FS group and negatively in PCs in a large tn cluster comprising the left UF and adjacent structures (cluster 1). We also identified negative correlations in FS and positive correlations in PCs between TNF-R1 and NDI in the UF and rin adjacent WM tracts (cluster 1). Other significant associations between NDI and serum biomarkers are shown in Tables 2 and 3, and Figure 1. ep ODI was differentially associated with several inflammatory biomarkers. Higher levels of TNF- were associated with lower ODI in FS and higher ODI in PCs in the left cingulum bundle and adjacent structures (cluster 3). Higher levels of ICAM-1 were associated with higher ODI in Pr FS and lower ODI in PCs in the left UF and adjacent structures (cluster 1). Other significant relationships between serum biomarkers and ODI are shown in Tables 2 and 3, and Figure 2. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed F-ISO was differentially associated with IL-6, TNF-R1, and TRAIL. IL-6 was negatively associated with F-ISO in FS and positively in PCs in the left UF and adjacent WM tracts (cluster 2). Higher TNF-R1 was associated with higher F-ISO in FS and lower F-ISO in PCs in the left UF and surrounding structures (cluster 1). Other associations between the serum biomarkers and FS PCs IL-6 NDI F-ISO NDI F-ISO TNF- ODI ODI TNF-R1 NDI F-ISO NDI F-ISO TRAIL NDI ODI NDI ODI ICAM-1 ODI ODI MCP-1 NDI F-ISO pe er r Serum biomarker ev F-ISO are shown in Tables 2 and 3. ot NDI Table 3. Associations between neurite density (NDI), orientation dispersion (ODI), fraction of tn isotropic diffusion (F-ISO), and serum biomarkers of inflammation in non-epileptic seizures (FS) and psychiatric controls (PCs). rin positive correlation; negative correlation (if both arrows present it indicates the relationship were either positive or negative depending on the brain region) ICAM-1=intercellular adhesion molecule 1, IL-6=interleukin 6, MCP-1=monocyte ep chemoattractant protein 1, TNF-=tumor necrosis factor , TNF-R1=tumor necrosis factor Pr receptor 1, TRAIL=tumor necrosis factor-related apoptosis inducing ligand This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed ev er r pe ot tn rin Figure 1. Clusters with significant interactions between group and serum biomarkers on neurite ep density. Dotted lines indicate linear trendlines. Images are shown in neurological convention (left=left). FS=functional seizures, IL-6=interleukin 6, MCP-1=monocyte chemoattractant protein 1, NDI=neurite density index, PC=psychiatric controls, TNF-R1=tumor necrosis factor Pr receptor 1, TRAIL= tumor necrosis factor-related apoptosis-inducing ligand This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed ev er r pe ot tn rin Figure 2. Clusters with significant interactions between group and serum biomarkers on neurite ep orientation dispersion. Dotted lines indicate linear trendlines. Images are shown in neurological convention (left=left). FS=functional seizures, ICAM-1=intercellular adhesion molecule 1, Pr ODI=orientation dispersion index, PC=psychiatric controls, TNF-=tumor necrosis factor , TRAIL= tumor necrosis factor-related apoptosis-inducing ligand This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Correlations specific to FS and PCs Next, we investigated bivariate associations between serum biomarkers and the NODDI metrics separately in FS patients and PCs. In FS, ICAM-1 was positively associated with NDI in the left CST and adjacent structures (cluster 2), and TNF-R1 was negatively associated with NDI ev in the left UF and adjacent structures. Other significant findings are listed in Table 4. TNF-, MCP-1, and TRAIL were not independently associated with NDI in the FS group. In PCs, TNF- er r R1 was positively associated with NDI in the right UF and adjacent structures (cluster 2). TNFR1 was also positively associated with NDI in the left anterior corona radiata, cingulum, forceps minor, and the genu and body of the corpus callosum (cluster 3). Other significant findings are pe listed in Table 4. There were no associations between NDI and ICAM-1, TNF-, or TRAIL in PCs. In FS, MCP-1 was positively associated with ODI in the left CST and adjacent structures ot (cluster 1) and in the right cingulum bundle and forceps minor (cluster 2). Other significant tn associations are reported in Table 4. In PCs, ICAM-1 was positively associated with ODI in the left CST and adjacent structures (cluster 6) and negatively associated with ODI in the left UF and adjacent structures (cluster 1). TNF-R1 was negatively correlated with ODI in the left CST and rin adjacent tracts (cluster 4) and positively correlated with ODI in the left UF and adjacent tracts (cluster 2). Other significant associations are reported in Table 4. ep In FS, ICAM-1 was negatively associated with F-ISO in the left CST and adjacent WM tracts (cluster 2). TNF-R1 was positively associated with F-ISO in the left UF and adjacent WM Pr tracts (cluster 1). In PCs, TNF-R1 was negatively associated with F-ISO in the right UF and This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed adjacent structures (cluster 2), and the cingulum bundle and adjacent structures (cluster 3). Other significant associations are reported in Table 4. MNI coordinates (peak signal) Hem. x y z Predictor and Region direction of association (+/-) TNF-R1 in FS cluster 1 (-) MCP-1 in PCs cluster 1 (-) 0 2172 R +28 +33 +16 1772 er r -48 -30 -15 +38 741 L -33 0 +39 735 L -33 +32 +15 1990 Unclassified WM adjacent to pre- and postcentral gyrus L -31 -20 +61 657 Ant. internal capsule Post. internal capsule Ant. corona radiata Sup. corona radiata Ant. thalamic radiation External capsule UF Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Cingulum bundle Ant. corona radiata L -33 -12 +39 4940 R +26 +25 +7 2065 L -11 +24 +22 1092 UF Ant. corona radiata Ant. thalamic radiation Inf. fronto-occipital fasciculus ep rin TNF-R1 in PCs cluster 1 (+) +40 L pe cluster 3 (+) Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Genu of corpus callosum CST Sup. corona radiata Sup. longitudinal fasciculus Unclassified WM adjacent to middle frontal and precentral gyrus R ot cluster 2 (+) Inf. fronto-occipital fasciculus tn IL-6 in FS cluster 1 (-) ICAM-1 in FS cluster 1 (+) ev NDI Cluster size (mm3) Pr cluster 2 (+) cluster 3 (+) This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
ODI cluster 2 (+) TRAIL in FS cluster 1 (+) TNF- in PCs cluster 1 (+) cluster 2 (+) 2395 L -19 -50 +20 949 Post. corona radiata Splenium of corpus callosum L -23 -49 +27 1166 -8 +2 889 CST Post. internal capsule Sup. corona radiata Post. corona radiata Cingulum bundle Forceps minor cluster 2 (+) ep cluster 3 (+) Pr cluster 4 (-) ev -3 L -23 R +11 +44 +25 831 Ant. thalamic radiation Inf. fronto-occipital fasciculus R +34 +51 -4 738 Sup. longitudinal fasciculus Splenium corpus callosum Body of corpus callosum Forceps major Sup. longitudinal fasciculus R L +26 -10 -48 -35 +59 +14 954 734 R +42 -58 +43 680 UF Ant. corona radiata Ant. internal capsule Ant. thalamic radiation Inf. fronto-occipital fasciculus Forceps minor Post. corona radiata Sup. corona radiata Sup. longitudinal fasciculus Splenium of corpus callosum Body of corpus callosum Post. corona radiata Forceps major Post. thalamic radiation Inf. fronto-occipital Inf. longitudinal fasciculi Forceps major L -21 +32 0 1913 R +27 -26 +30 1615 L -4 -36 +8 1284 L -35 -60 -1 915 rin cluster 3 (+) ICAM-1 in PCs cluster 1 (-) +49 er r MCP-1 in FS cluster 1 (+) +38 pe ICAM-1 in FS cluster 1 (+) R ot cluster 2 (+) Unspecified WM adjacent to inf. fronto-occipital gyrus Splenium of corpus callosum Forceps major tn IL-6 in FS cluster 1 (-) iew ed Forceps minor Genu of corpus callosum Body of corpus callosum This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
cluster 2 (+) cluster 3 (+) +31 +8 878 833 R +16 -62 +52 768 R +35 +34 +10 679 Ant. internal capsule Post. internal capsule Sup. corona radiata Sup. fronto-occipital fasciculus Ant. thalamic radiation UF Genu of corpus callosum Ant. corona radiata Ant. thalamic radiation Inf. fronto-occipital fasciculus Forceps minor Ant. internal capsule Sup. corona radiata Sup. fronto-occipital fasciculus Ant. thalamic radiation CST Post. internal capsule Retrolenticular internal capsule Post. corona radiata Sup. corona radiata External capsule F-ISO L -20 -9 +20 1469 L -23 +37 +2 1445 R +20 +17 +11 1012 L -27 -22 +14 805 Unclassified WM adjacent to ant. thalamic radiation and inf. frontooccipital fasciculus R +21 +60 -14 3308 Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus Genu of corpus callosum CST Sup. corona radiata Sup. longitudinal fasciculus Ant. corona radiata R +29 +32 +15 1585 L -30 -14 +37 919 L -32 +28 +15 883 rin IL-6 in FS cluster 1 (-) tn ot cluster 4 (-) -25 -25 iew ed TNF-R1 in PCs cluster 1 (+) +42 -27 ev cluster 8 (+) R L er r cluster 7 (+) Sup. longitudinal fasciculus CST Retrolenticular internal capsule Post. corona radiata Unclassified WM adjacent to sup. lateral occipital cortex and sup. parietal lobe Ant. thalamic radiation Inf. fronto-occipital fasciculus pe cluster 5 (-) cluster 6 (+) ep ICAM-1 in FS cluster 1 (-) Pr cluster 2 (-) cluster 3 (-) This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
TNF-R1 in FS cluster 1 (+) UF Ant. corona radiata Ant. thalamic radiation Inf. fronto-occipital fasciculus TNF-R1 in PCs cluster 1 (-) L iew ed Ant. thalamic radiation Inf. fronto-occipital fasciculus -32 +28 +15 1936 ot pe er r ev Ant. internal capsule L -32 -12 +42 3408 Ant. corona radiata Sup. corona radiata Ant. thalamic radiation External capsule Inf. fronto-occipital faciculus cluster 2 (-) UF R +21 +36 +9 2109 Ant. corona radiata Ant. thalamic radiation Forceps minor Inf. fronto-occipital fasciculus cluster 3 (-) Cingulum bundle L -19 +35 +8 1109 Genu of corpus callosum Body of corpus callosum Anterior corona radiata Anterior thalamic radiation Forceps minor Table 4. Clusters with significant associations between NDI, ODI, F-ISO and serum biomarkers in FS and PCs. tn Note: Only tracts with cluster overlap of >3% are reported. Ant.=anterior, CST=corticospinal tract, F-ISO=fraction of isotropic diffusion, FS=functional rin seizures, Hem.=hemisphere, ICAM-1=intercellular adhesion molecule 1, IL-6=interleukin 6, Inf.=inferior, L=left, MCP-1=monocyte chemoattractant protein 1, NDI=neurite density index, MNI=Montreal Neurological Institute, ODI=orientation dispersion index, PCs=psychiatric ep controls, Post.=posterior, R=right, Sup.=superior, TNF-=tumor necrosis factor , TNFR1=tumor necrosis factor receptor 1, TRAIL=tumor necrosis factor-associated apoptosis- Pr inducing ligand, UF=uncinate fasciculus, WM=white matter This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Correlations between serum biomarkers and clinical outcomes Significant correlations between serum biomarkers and clinical outcomes (questionnaire scores) are shown in Figure 3. There were no significant correlations (at p<0.0126) between IL6, MCP-1, TRAIL, and any of the clinical outcomes. Higher TNF- was associated with lower medication-related QoL on the QOLIE-31 in ev the FS group (r=-0.670, p<0.001). Higher ICAM-1 was associated with higher interpersonal sensitivity (r=0.603, p=0.003), hostility (r=0.549, p=0.008), and global SCL-90 score (r=0.527, er r p=0.012) in FS. ICAM-1 was also positively associated with the Life Activities (r=0.650, p=0.001) and Participation subscales (r=0.630, p=0.002) of the WHODAS2.0, and with total WHODAS2.0 disability scores (r=0.542, p=0.009) in FS, indicating higher levels of disability pe with increasing ICAM-1. There were no such correlations in PCs. Finally, TNF-R1 was positively associated with HADS depression (r=0.550, p=0.008) and anxiety (r=0.554, p=0.007) and negatively associated with emotion-related QoL (r=-0.647, p=0.001) in FS. Higher TNF-R1 ot was also associated with greater disability related to cognitive functioning (r=0.641, p=0.001), getting along with others (r=0.569, p=0.006), participation (r=0.642, p=0.001), and with total tn disability (r=0.628, p=0.002) in FS. These associations were not present in PCs. There were no associations between serum biomarkers and one-week FS count rin (Spearman correlation coefficient) in the FS group. There were no associations with stressful life Pr ep events, dissociative experiences, or somatoform dissociation in either group. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed ev er r pe ot tn Figure 3. Associations between serum biomarkers and clinical outcomes in functional seizures (FS, solid circles) and psychiatric controls (PCs, empty circles). Correlations are significant at rin p<0.0126 (adjusted for multiple comparisons) in FS, and not significant in PCs. Trendlines show ep linear relationships in FS (solid line) and PCs (dotted line). Discussion Pr The main aim of this study was to assess WM microstructure in FS compared to PCs and its relationship to peripheral biomarkers of inflammation. We did not find differences between This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed FS WM microstructure compared to controls with MHDs. However, as expected, we identified several group differences in the relationship between peripheral inflammatory biomarkers and NODDI metrics in the UF, cingulum bundle, and CST, as well as significant associations between serum biomarkers and clinical symptoms. The lack of group differences in NODDI metrics is the first important finding in this ev study. As mentioned above, several studies have compared diffusion-based metrics between FS and HCs using DTI, and one used NODDI in individuals with FS and history of TBI. In the only er r study that used NODDI in FS, Goodman et al. 18 reported decreased NDI, F-ISO, and FA and increased MD in the cingulum bundle, UF, FST, and CST, and we expected to replicate these findings. However, as we did not find the expected differences, we believe this null finding is pe very important as it indicates that differences in sample characteristics (individuals with prior TBI versus general FS population) and lack of control for comorbid MHDs may explain the discrepant findings. 53 ot All other studies of WM integrity in FS have focused on comparisons with HCs. Hernando et al. 15 compared UF integrity between eight individuals with FS and eight matched tn HCs. While the FS group showed more rightward asymmetry in the UF than controls as indicated by the number of streamlines on DTI tractography, FA did not differ between groups. rin Although their sample size was small, their findings are consistent with the current results, indicating no impaired WM integrity in the UF in FS compared to HCs. Conversely, Lee et al. ep reported increased FA in the left UF of FS patients compared to HCs, indicating greater WM integrity. No psychiatric comorbidities or correlations with clinical symptoms were reported, Pr thus, it is not possible to determine if the UF differences were related to FS or to underlying comorbid psychiatric comorbidities. Jungilligens et al. 16 also reported a lack of differences in FA This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed values throughout the brain between 20 FS and 20 HCs. Lower FA in the UF, FST, and corpus callosum was related to impaired performance on an emotion processing task, earlier age of symptom onset, and higher FS frequency, indicating that UF integrity has important clinical implications. A leftward as opposed to rightward UF asymmetry on DTI tractography was also found in FS. The authors acknowledged that because of significant psychiatric comorbidities in ev the FS group, it was not possible to determine whether the observed relationships were specific to FS or to psychopathology in general. Finally, Sone et al. (2019) 54 reported decreased FA and er r increased mean diffusivity (MD), indicating impaired WM integrity throughout the WM in 17 patients with FS. The affected tracts were not further detailed, so it is not possible to compare their findings to the current study. The authors also recognized that psychiatric comorbidities pe were a significant confounding factor in interpreting the findings, although no further attempt was made to address this. In summary, because of the uncertain contributions from MHDs and neurological comorbidities in prior FS studies, and our current findings of no WM differences ot between FS and PCs, we suspect that previously identified WM abnormalities in FS are connected to comorbid MHDs or specific psychological symptoms commonly experienced by FS tn patients rather than constituting FS-specific pathology. This notion is further supported by the fact that abnormalities in the UF and other WM tracts have been previously reported in major rin depressive disorder (MDD), 55-57 generalized anxiety disorder, 57 and PTSD, 58-61 with one study suggesting that UF abnormalities may be related to dissociative symptoms in PTSD. 60 It is clear ep that identifying a specific neurological signature of FS must involve comparisons with psychiatric conditions involving symptoms similar to FS. Pr When analyzing the relationships between serum biomarkers of inflammation and the NODDI metrics, the most prominent associations were found in the UF, which is a WM tract that This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed connects the medial temporal with the medial frontal lobes. 62 Among other functions, the UF is thought to allow individuals to draw from prior experience (memory) when making decisions and it is known to take part in emotion regulation. 62,63 DMRI abnormalities in the UF have been observed in FS/FNSD studies14,15,19 but, as mentioned above, they may be related to the presence of comorbid mental health disorders rather than specific to FS. In our FS group, higher IL-6 was ev associated with better WM integrity (higher NDI and lower F-ISO) in a largely overlapping cluster comprising the left UF and adjacent WM (Figure 4). Conversely, in PCs, IL-6 was er r associated with lower NDI and higher F-ISO, as expected based on prior literature that has pointed to IL-6 in the pathophysiology of depression. 28 IL-6 is a pro-inflammatory cytokine released by various immune cells in response to injury and infection. It has important immune pe functions, such as inducing immune cell growth and differentiation, and potentiating inflammatory responses. 64,65 Chronic IL-6 overproduction has been linked to the development of Pr ep rin tn ot autoimmune diseases and certain cancers66 and to MDD. 30 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed ev er r pe ot Figure 4. Clusters with significant interactions between group and interleukin-6. NDI only tn Blue=interactions on F-ISO only, Purple=interactions on NDI and F-ISO, Red=interactions on rin Our findings in the PC group are consistent with the inflammatory hypothesis of depression28,67 and suggest that compromised WM integrity in the UF may be one link between ep IL-6 and depressive symptoms. This is plausible because IL-6 can cross the blood-brain barrier and induce neurodegeneration. 68-70 Our results also indicate that detrimental relationships Pr between IL-6 and UF integrity may distinguish depression from FS. We note that our observations are correlative, so it is not possible to determine whether IL-6 caused damage to the This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed UF. Further, serum IL-6 was not significantly higher in PCs than FS, and was not associated with clinical outcomes. Because our control group was partly composed of individuals with anxiety and trauma-related disorders, existing relationships between IL-6 and clinical symptoms may have been concealed by the heterogeneity of the control group. In contrast to the relationship between IL-6 and UF in PCs, TNF-R1 was associated with ev UF pathology in FS. In FS, higher levels of TNF-R1 were associated with lower UF NDI and higher UF F-ISO (Figure 5), with the opposite relationships observed in PCs. TNF-R1 is er r expressed on immune cells including microglia and macrophages and is involved in cellular apoptosis, survival, differentiation, and inflammation. 71 Because TNF-R1 binding exacerbates inflammation, elevated serum TNF-R1 is used as a marker of active inflammation. TNF-R1 was pe the only inflammatory biomarker that was significantly elevated in the FS group, and increased TNF-R1 was associated with higher depression and anxiety, and worse QoL, cognitive functioning, and disability. We initially expected that TNF-R1 would be elevated in PCs ot compared to FS because it was previously linked to bipolar disorder. 36 Because in the current study PCs included individuals with depression but not bipolar disorder, this may explain the tn discrepant findings. Together, our results suggest that while both FS and other psychiatric conditions involve inflammation, the effects of this on WM integrity may be dependent to the rin particular inflammatory pathways activated, with TNF-R1 related to UF pathology in FS, and IL- Pr ep 6 related to UF pathology in PCs. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed ev er r pe Figure 5. Clusters with significant interactions between group and tumor necrosis factor receptor 1. Blue=interactions on F-ISO only, Purple=interactions on NDI and F-ISO, Red=interactions on tn ot NDI only Another WM tract affected by serum biomarker levels was the CST. The CST is an rin efferent WM tract originating in cortical motor areas with a main role in the control of voluntary motor activities, which may be affected during FS if patients are experiencing convulsions, stiffness, and weakness. 72 The CST was chosen as a tract of interest for distinguishing FS from ep PCs due to the prominent motor symptoms of FS and prior literature showing CST abnormalities in FS. 18 We found no differences in CST WM microstructure between FS and PCs, which is an Pr important finding that suggests CST integrity could be a transdiagnostic correlate of motor symptoms in psychiatric disorders. This is plausible given that motor symptoms such as This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed psychomotor slowing and agitation are also prominent in MHDs. 73-76 Despite the lack of overall differences, the two groups were characterized by different serum biomarker profiles with respect to CST integrity. In FS, higher ICAM-1 was associated with higher NDI and lower F- ISO in the CST, indicating higher tract integrity and suggesting that ICAM-1 is not a risk factor for CST damage in FS. However, higher ICAM-1 was associated with higher levels of disability ev and psychological symptoms in FS, suggesting that ICAM-1 may be related to FS symptoms via other mechanisms. er r Lower MCP-1 was associated with lower ODI in the CST of FS patients. ODI reflects the complexity of the underlying WM structure; areas with crossing fibers and dendritic arborizations and the gray matter have higher ODI, whereas areas with parallel axons, such as pe major WM bundles, have lower ODI. In disease, increased ODI in the gray matter has been associated with regrowth and reorganization, 77 so it may be particularly useful for assessing therapeutic response in longitudinal studies. In the WM, increased ODI indicates axonal ot degeneration and demyelination. 13 This means that individuals with FS and lower MCP-1 levels were more likely to show CST damage in our study. MCP-1 levels did not correlate with any tn clinical outcomes. In PCs, decreased ICAM-1 and increased TNF-R1 were associated with lower CST ODI, rin which points to TNF-R1 as a risk factor for CST abnormalities in this group, although because of the lack of clinical correlations, the potential effects of such changes are unclear. ep The final brain region to be affected by serum biomarkers was the cingulum bundle, particularly in PCs, where higher TNF-R1 was associated with higher NDI and lower F-ISO. The Pr cingulum bundle connects the medial temporal with the frontal and parietal lobes and subcortical nuclei with the cingulate cortex. It supports executive functioning, emotion and pain regulation, This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed and memory, and structural and functional abnormalities in the cingulum bundle have been implicated in depression, 78,79 PTSD, 80,81 and FNSDs. 82 Our findings show that PCs with higher levels of serum TNF-R1 show higher cingulum bundle integrity than PCs with lower TNF-R1. Because TNF-R1 levels were not related to clinical symptoms in PCs, their clinical relevance is unclear, but they are consistent with prior literature implicating the cingulum in disorders ev presenting with mood symptoms. 83 We found associations between serum biomarkers and NODDI metrics in many other er r WM tracts. These results are reported to guide the selection of brain regions for further study of FS neuropathophysiology. The only association with TRAIL in FS was found in the right anterior thalamic radiation and inferior fronto-occipital fasciculus, which were not tracts of pe interest. TRAIL was also not related to any clinical symptoms in the FS group. Because TRAIL appears to be elevated immediately after FS, 2 this biomarker should remain a target for future studies. ot Finally, we note the relationships between three of the six inflammatory biomarkers (TNF-, TNF-R1, and ICAM-1) and disability, QoL, and psychological problems including tn anxiety and depression in FS. TNF-R1 showed the most prominent associations with clinical symptoms, including with higher depression, anxiety, lower emotional QoL, and higher levels of rin disability. TNF-R1 was related to decreased F-ISO in the cingulum bundle and to lower NDI and higher F-ISO in the UF in FS, suggesting that these areas may be particularly relevant to FS ep symptoms. Symptoms specific to FS, such as somatoform dissociation, were not related to any serum biomarkers tested in the current study. We suggest that future studies assess a broader Pr range of serum biomarkers of inflammation to determine whether any may be related to dissociative symptoms. Because dissociative symptoms are central to FS, PTSD and dissociative This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed disorders, candidate biomarkers for FS should be able to distinguish between patients with these conditions. There were no significant associations between serum biomarkers and any clinical symptoms in PCs. This was unexpected given that elevated levels of several of the tested biomarkers have previously been identified in MDD. 67,84-88 However, the lack of significant ev associations may be due to differences in measures in the current study, which were primarily selected to capture FS symptomatology, due to heterogeneity of MDDs in both groups, or to er r treatment effects. The elevated TNF-R1 levels in the FS group alongside the lack of WM abnormalities when compared to PCs provide support for the two-hit hypothesis of FS, which suggests that pe traumatic experiences prime the brain toward NI when trauma is subsequently experienced later in life. We did observe that higher serum TNF-R1 was associated with UF pathology in FS, but the overall lack of UF abnormalities when compared to PCs suggests that either inflammation is ot not severe enough to damage WM integrity in FS, or that relationships between traumatic life experiences, inflammation, and WM damage are not unique to FS. The similar levels of trauma tn exposure in our two groups suggest that the latter may be the case. However, we also note that inflammatory biomarkers in serum are not a direct measure of NI, and animal models of FS that rin would allow us to quantify inflammatory cytokines in the brain have not been developed. To test the two-hit hypothesis in humans more directly, longitudinal monitoring of FS patients with ep neuroimaging could be used to determine the relative timing of traumatic experiences, NI, and Pr FS symptoms. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Limitations There are several limitations to the current study that should be considered. First, our FS group was recruited from an outpatient specialty clinic and all patients were undergoing, or had recently completed, psychological treatment for FS. Psychotherapeutic interventions such as mindfulness have been shown to affect immune system functioning, including levels of ev circulating cytokines, in patients with anxiety and depression. 89-91 Thus, it is possible that untreated patients with FS would show different relationships between inflammatory cytokines, er r neuroimaging markers, and clinical symptoms than the current sample. Because most FS participants were experiencing few FS each week (average=2/week; Table 1), this may also have affected our ability to detect expected associations between inflammatory biomarkers and FS pe frequency. Further, we did not control for the therapeutic approaches used as participants received personalized interventions as appropriate for their presentation. 92 The specific effects of these interventions on WM and immunity are not known and need to be investigated. ot We did not time the serum collection in relation to FS. It is possible that there is a relationship between inflammatory biomarkers and time since last FS. For example, Gledhill et tn al. 2 described cytokine elevations within 24 hours of FS and epileptic seizures. Future studies may account for time since the most recent FS when assessing relationships between rin inflammatory biomarkers and neuroimaging outcomes. Lastly, our control group was comprised of individuals with various MHDs. We recruited ep participants with a range of psychological diagnoses to better reflect comorbid symptoms in FS that could account for neuroimaging and biomarker abnormalities in FS. By increasing the Pr specificity of our findings for FS, we may have lost some sensitivity for detecting differences between FS and each of these diagnoses. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Conclusion For the first time, we report relationships between peripheral inflammatory biomarkers and WM abnormalities and symptom severity specific to FS. The UF, CST, and cingulum bundle emerged as relevant areas when distinguishing FS from PCs. Once our findings are replicated in ev future studies, specific serum biomarkers related to inflammation could be used in clinical settings to aid FS diagnosis and to quantify treatment response, with TNF-R1 in particular being er r a promising candidate for further study. This may be particularly useful in settings where videoEEG is not available. The lack of group differences in WM microstructure suggests that previously identified WM abnormalities in FS versus HCs may in part be related to the pe psychological symptoms experienced by FS patients. ot Acknowledgments We would like to thank Dr. Christopher Litton and Holly Yazdi for assistance with tn recruitment and data collection, respectively. Declaration of interest rin JPS is funded by NIH, DoD, State of Alabama, Shor Foundation for Epilepsy Research, UCB Pharma Inc., and NeuroPace Inc. Has served on consulting/advisory boards for Greenwich ep Biosciences Inc., NeuroPace, Inc., Serina Therapeutics Inc., LivaNova Inc., UCB Pharma Inc., iFovea, AdCel Biopharma LLC, and Elite Medical Experts LLC. He serves as an editorial board Pr member for Epilepsy & Behavior, Journal of Epileptology (associate editor), Epilepsy & This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
iew ed Behavior Reports (Editor-in-Chief), Journal of Medical Science, Epilepsy Currents (contributing editor), and Folia Medica Copernicana. Funding This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. This study was supported by start-up funds provided to JPS ev by the Department of Neurology, UAB Heersink School of Medicine. The sponsor was not involved in the study design; in the collection, analysis and interpretation of data; in the writing er r of the report; and in the decision to submit the article for publication. References Sharma AA, Szaflarski JP. Neuroinflammation as a pathophysiological factor in the pe 1. development and maintenance of functional seizures: A hypothesis. Epilepsy Behav Rep. 2021;16:100496. doi:10.1016/j.ebr.2021.100496 Gledhill JM, Brand EJ, Pollard JR, St Clair RD, Wallach TM, Crino PB. Association of ot 2. Epileptic and Nonepileptic Seizures and Changes in Circulating Plasma Proteins Linked to tn Neuroinflammation. Neurology. Mar 9 2021;96(10):e1443-e1452. doi:10.1212/wnl.0000000000011552 Paredes-Echeverri S, Maggio J, Bègue I, Pick S, Nicholson TR, Perez DL. Autonomic, rin 3. Endocrine, and Inflammation Profiles in Functional Neurological Disorder: A Systematic ep Review and Meta-Analysis. J Neuropsychiatry Clin Neurosci. Winter 2022;34(1):30-43. doi:10.1176/appi.neuropsych.21010025 4. Nehring SM, Goyal A, Patel BC. C Reactive Protein. StatPearls. StatPearls Publishing Pr Copyright © 2022, StatPearls Publishing LLC.; 2022. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Kozlowska K, Chung J, Cruickshank B, et al. Blood CRP levels are elevated in children iew ed 5. and adolescents with functional neurological symptom disorder. European Child & Adolescent Psychiatry. 2019/04/01 2019;28(4):491-504. doi:10.1007/s00787-018-1212-2 6. Hanly JG, Kozora E, Beyea SD, Birnbaum J. Review: Nervous System Disease in Systemic Lupus Erythematosus: Current Status and Future Directions. Arthritis & rheumatology 7. Mackay M, Tang CC, Vo A. Advanced neuroimaging in neuropsychiatric systemic lupus doi:10.1097/wco.0000000000000822 er r erythematosus. Curr Opin Neurol. Jun 2020;33(3):353-361. 8. ev (Hoboken, NJ). Jan 2019;71(1):33-42. doi:10.1002/art.40591 Rana A, Musto AE. The role of inflammation in the development of epilepsy. Journal of 9. pe neuroinflammation. May 15 2018;15(1):144. doi:10.1186/s12974-018-1192-7 Cunningham C. Microglia and neurodegeneration: the role of systemic inflammation. Glia. Jan 2013;61(1):71-90. doi:10.1002/glia.22350 Paredes-Echeverri S, Maggio J, Bègue I, Pick S, Nicholson TR, Perez DL. Autonomic, ot 10. Endocrine, and Inflammation Profiles in Functional Neurological Disorder: A Systematic tn Review and Meta-Analysis. The Journal of Neuropsychiatry and Clinical Neurosciences. 2022/02/01 2021;34(1):30-43. doi:10.1176/appi.neuropsych.21010025 Sepehrband F, Clark KA, Ullmann JF, et al. Brain tissue compartment density estimated rin 11. using diffusion-weighted MRI yields tissue parameters consistent with histology. Hum Brain ep Mapp. Sep 2015;36(9):3687-702. doi:10.1002/hbm.22872 12. Wang ZX, Zhu WZ, Zhang S, Shaghaghi M, Cai KJ. Neurite Orientation Dispersion and Pr Density Imaging of Rat Brain Microstructural Changes due to Middle Cerebral Artery Occlusion at a 3T MRI. Curr Med Sci. Feb 2021;41(1):167-172. doi:10.1007/s11596-021-2332-3 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Grussu F, Schneider T, Tur C, et al. Neurite dispersion: a new marker of multiple iew ed 13. sclerosis spinal cord pathology? Annals of clinical and translational neurology. Sep 2017;4(9):663-679. doi:10.1002/acn3.445 14. Lee S, Allendorfer JB, Gaston TE, et al. White matter diffusion abnormalities in patients with psychogenic non-epileptic seizures. Brain research. Sep 16 2015;1620:169-76. 15. ev doi:10.1016/j.brainres.2015.04.050 Hernando KA, Szaflarski JP, Ver Hoef LW, Lee S, Allendorfer JB. Uncinate fasciculus er r connectivity in patients with psychogenic nonepileptic seizures: A preliminary diffusion tensor tractography study. Epilepsy & Behavior. 2015/04/01/ 2015;45:68-73. doi:https://doi.org/10.1016/j.yebeh.2015.02.022 Jungilligens J, Wellmer J, Kowoll A, Schlegel U, Axmacher N, Popkirov S. pe 16. Microstructural integrity of affective neurocircuitry in patients with dissociative seizures is associated with emotional task performance, illness severity and trauma history. Seizure. Jan 17. ot 2021;84:91-98. doi:10.1016/j.seizure.2020.11.021 Ding JR, An D, Liao W, et al. Altered functional and structural connectivity networks in tn psychogenic non-epileptic seizures. PloS one. 2013;8(5):e63850. doi:10.1371/journal.pone.0063850 Goodman AM, Allendorfer JB, Blum AS, et al. White matter and neurite morphology rin 18. differ in psychogenic nonepileptic seizures. https://doi.org/10.1002/acn3.51198. Annals of ep clinical and translational neurology. 2020/10/01 2020;7(10):1973-1984. Pr doi:https://doi.org/10.1002/acn3.51198 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Perez DL, Nicholson TR, Asadi-Pooya AA, et al. Neuroimaging in Functional iew ed 19. Neurological Disorder: State of the Field and Research Agenda. NeuroImage: Clinical. 2021/01/01/ 2021;30:102623. doi:https://doi.org/10.1016/j.nicl.2021.102623 20. Szaflarski JP, Allendorfer JB, Nenert R, et al. Facial emotion processing in patients with seizure disorders. Epilepsy & behavior : E&B. Feb 2018;79:193-204. 21. ev doi:10.1016/j.yebeh.2017.12.004 Rahimian R, Belliveau C, Chen R, Mechawar N. Microglial Inflammatory-Metabolic er r Pathways and Their Potential Therapeutic Implication in Major Depressive Disorder. Front Psychiatry. 2022;13:871997. doi:10.3389/fpsyt.2022.871997 22. Wohleb ES. Neuron-Microglia Interactions in Mental Health Disorders: "For Better, and 23. pe For Worse". Front Immunol. 2016;7:544. doi:10.3389/fimmu.2016.00544 Szaflarski JP. The Molecular Dichotomy Between Epileptic and Functional Seizures. Epilepsy Currents. 2021/07/01 2021;21(4):258-260. doi:10.1177/15357597211011983 Szaflarski JP, LaFrance WC, Jr. Psychogenic Nonepileptic Seizures (PNES) as a ot 24. Network Disorder - Evidence From Neuroimaging of Functional (Psychogenic) Neurological 25. tn Disorders. Epilepsy Curr. Jul-Aug 2018;18(4):211-216. doi:10.5698/1535-7597.18.4.211 Oestreich LKL, O’Sullivan MJ. Transdiagnostic In Vivo Magnetic Resonance Imaging rin Markers of Neuroinflammation. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging. 2022/07/01/ 2022;7(7):638-658. doi:https://doi.org/10.1016/j.bpsc.2022.01.003 Vavasour IM, Sun P, Graf C, et al. Characterization of multiple sclerosis ep 26. neuroinflammation and neurodegeneration with relaxation and diffusion basis spectrum imaging. Pr Mult Scler. Mar 2022;28(3):418-428. doi:10.1177/13524585211023345 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Hutchinson EB, Schwerin SC, Avram AV, Juliano SL, Pierpaoli C. Diffusion MRI and iew ed 27. the detection of alterations following traumatic brain injury. J Neurosci Res. Apr 2018;96(4):612-625. doi:10.1002/jnr.24065 28. Roohi E, Jaafari N, Hashemian F. On inflammatory hypothesis of depression: what is the role of IL-6 in the middle of the chaos? Journal of neuroinflammation. Feb 16 2021;18(1):45. 29. ev doi:10.1186/s12974-021-02100-7 Pae CU. The potential role of monocyte chemoattractant protein-1 for major depressive 30. Ting EY, Yang AC, Tsai SJ. Role of Interleukin-6 in Depressive Disorder. Int J Mol Sci. Mar 22 2020;21(6)doi:10.3390/ijms21062194 O'Brien SM, Scully P, Scott LV, Dinan TG. Cytokine profiles in bipolar affective pe 31. er r disorder. Psychiatry Investig. Jul 2014;11(3):217-22. doi:10.4306/pi.2014.11.3.217 disorder: focus on acutely ill patients. Journal of affective disorders. Feb 2006;90(2-3):263-7. doi:10.1016/j.jad.2005.11.015 Uzzan S, Azab AN. Anti-TNF-α Compounds as a Treatment for Depression. Molecules. ot 32. Apr 19 2021;26(8)doi:10.3390/molecules26082368 Berthold-Losleben M, Himmerich H. The TNF-alpha system: functional aspects in tn 33. depression, narcolepsy and psychopharmacology. Curr Neuropharmacol. Sep 2008;6(3):193- 34. rin 202. doi:10.2174/157015908785777238 Alshammari MA, Khan MR, Majid Mahmood H, et al. Systemic TNF-α blockade ep attenuates anxiety and depressive-like behaviors in db/db mice through downregulation of inflammatory signaling in peripheral immune cells. Saudi Pharm J. May 2020;28(5):621-629. Pr doi:10.1016/j.jsps.2020.04.001 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Dib P, Zhang Y, Ihnat MA, Gallucci RM, Standifer KM. TNF-Alpha as an Initiator of iew ed 35. Allodynia and Anxiety-Like Behaviors in a Preclinical Model of PTSD and Comorbid Pain. Original Research. Frontiers in Psychiatry. 2021-August-25 2021;12doi:10.3389/fpsyt.2021.721999 36. Soczynska JK, Kennedy SH, Goldstein BI, Lachowski A, Woldeyohannes HO, McIntyre ev RS. The effect of tumor necrosis factor antagonists on mood and mental health-associated quality of life: novel hypothesis-driven treatments for bipolar depression? Neurotoxicology. Jul 37. er r 2009;30(4):497-521. doi:10.1016/j.neuro.2009.03.004 Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. Jun 2009;29(6):313-26. 38. pe doi:10.1089/jir.2008.0027 LaFrance WC, Jr., Baker GA, Duncan R, Goldstein LH, Reuber M. Minimum requirements for the diagnosis of psychogenic nonepileptic seizures: a staged approach: a report ot from the International League Against Epilepsy Nonepileptic Seizures Task Force. Epilepsia. Nov 2013;54(11):2005-18. doi:10.1111/epi.12356 Stern AF. The Hospital Anxiety and Depression Scale. Occupational Medicine. tn 39. 2014;64(5):393-394. doi:10.1093/occmed/kqu024 Carlson EB, Putnam FW. An update on the dissociative experiences scale. Dissociation: rin 40. progress in the dissociative disorders. 1993; Schnurr P, Vielhauer M, Weathers F, Findler M. Brief Trauma Questionnaire. National ep 41. Center for PTSD; 1999. Pr doi:https://www.ptsd.va.gov/professional/assessment/documents/BTQ.pdf This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Holmes TH, Rahe RH. The social readjustment rating scale. Journal of psychosomatic iew ed 42. research. 1967;11(2):213-218. 43. Derogatis LR. SCL-90-R: Administration, scoring and procedures manual. 3rd ed. NCS Pearson; 1994. 44. Nijenhuis ER, Spinhoven P, Van Dyck R, Van der Hart O, Vanderlinden J. The ev development and psychometric characteristics of the Somatoform Dissociation Questionnaire (SDQ-20). J Nerv Ment Dis. Nov 1996;184(11):688-94. doi:10.1097/00005053-199611000- 45. er r 00006 Cramer JA, Perrine K, Devinsky O, Bryant-Comstock L, Meador K, Hermann B. Development and Cross-Cultural Translations of a 31-Item Quality of Life in Epilepsy pe Inventory. https://doi.org/10.1111/j.1528-1157.1998.tb01278.x. Epilepsia. 1998/01/01 1998;39(1):81-88. doi:https://doi.org/10.1111/j.1528-1157.1998.tb01278.x 46. Üstün TB, Kostanjsek N, Chatterji S, Rehm J. Measuring health and disability: Manual 47. ot for WHO disability assessment schedule WHODAS 2.0. World Health Organization; 2010. Cox RW. AFNI: software for analysis and visualization of functional magnetic resonance 48. tn neuroimages. Comput Biomed Res. Jun 1996;29(3):162-73. doi:10.1006/cbmr.1996.0014 Cox RW, Hyde JS. Software tools for analysis and visualization of fMRI data. NMR rin Biomed. Jun-Aug 1997;10(4-5):171-8. doi:10.1002/(sici)1099-1492(199706/08)10:4/5<171::aidnbm453>3.0.co;2-l Irfanoglu MO, Nayak A, Jenkins J, Pierpaoli C. TORTOISE v3:Improvements and New ep 49. Features of the NIH Diffusion MRI Processing Pipeline. presented at: ISMRM 25th Annual Pr meeting; 2017; Honolulu, HI. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Pierpaoli C, Walker L, Irfanoglu M, et al. TORTOISE: an integrated software package iew ed 50. for processing of diffusion MRI data. 2010: 51. Irfanoglu MO, Modi P, Nayak A, Hutchinson EB, Sarlls J, Pierpaoli C. DR-BUDDI (Diffeomorphic Registration for Blip-Up blip-Down Diffusion Imaging) method for correcting echo planar imaging distortions. NeuroImage. 2015/02/01/ 2015;106:284-299. 52. ev doi:https://doi.org/10.1016/j.neuroimage.2014.11.042 Storey JD. A direct approach to false discovery rates. https://doi.org/10.1111/1467- er r 9868.00346. Journal of the Royal Statistical Society: Series B (Statistical Methodology). 2002/08/01 2002;64(3):479-498. doi:https://doi.org/10.1111/1467-9868.00346 53. Wallace EJ, Mathias JL, Ward L. Diffusion tensor imaging changes following mild, pe moderate and severe adult traumatic brain injury: a meta-analysis. Brain imaging and behavior. Dec 2018;12(6):1607-1621. doi:10.1007/s11682-018-9823-2 54. Sone D, Sato N, Ota M, Kimura Y, Matsuda H. Widely Impaired White Matter Integrity ot and Altered Structural Brain Networks in Psychogenic Non-Epileptic Seizures. Neuropsychiatr Dis Treat. 2019;15:3549-3555. doi:10.2147/ndt.S235159 Pfarr JK, Brosch K, Meller T, et al. Brain structural connectivity, anhedonia, and tn 55. phenotypes of major depressive disorder: A structural equation model approach. Hum Brain 56. rin Mapp. Oct 15 2021;42(15):5063-5074. doi:10.1002/hbm.25600 Duan J, Wei Y, Womer FY, et al. Neurobiological substrates of major psychiatry ep disorders: transdiagnostic associations between white matter abnormalities, neuregulin 1 and clinical manifestation. J Psychiatry Neurosci. Sep 1 2021;46(5):E506-e515. Pr doi:10.1503/jpn.200166 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Luttenbacher I, Phillips A, Kazemi R, et al. Transdiagnostic role of glutamate and white iew ed 57. matter damage in neuropsychiatric disorders: A Systematic Review. J Psychiatr Res. Mar 2022;147:324-348. doi:10.1016/j.jpsychires.2021.12.042 58. Kritikos M, Huang C, Clouston SAP, et al. DTI Connectometry Analysis Reveals White Matter Changes in Cognitively Impaired World Trade Center Responders at Midlife. J 59. ev Alzheimers Dis. 2022;89(3):1075-1089. doi:10.3233/jad-220255 Lan QY, Cao ZH, Qi RF, et al. [A study on longitudinal changes in white matter er r microstructure of parents who have lost their only child based on diffusion tensor imaging and its relationship with symptoms of posttraumatic stress disorder]. Zhonghua Yi Xue Za Zhi. Jun 21 2022;102(23):1760-1765. doi:10.3760/cma.j.cn112137-20211213-02778 Wiingaard Uldall S, Lundell H, Baaré WFC, Roman Siebner H, Rostrup E, Carlsson J. pe 60. White matter diffusivity and its correlations to state measures of psychopathology in male refugees with posttraumatic stress disorder. Neuroimage Clin. 2022;33:102929. 61. ot doi:10.1016/j.nicl.2021.102929 Suo X, Lei D, Li W, et al. Psychoradiological abnormalities in treatment-naive tn noncomorbid patients with posttraumatic stress disorder. Depress Anxiety. Jan 2022;39(1):83-91. doi:10.1002/da.23226 Von Der Heide RJ, Skipper LM, Klobusicky E, Olson IR. Dissecting the uncinate rin 62. fasciculus: disorders, controversies and a hypothesis. Brain. Jun 2013;136(Pt 6):1692-707. ep doi:10.1093/brain/awt094 63. Goodman AM, Harnett NG, Knight DC. Pavlovian conditioned diminution of the Pr neurobehavioral response to threat. Neuroscience and biobehavioral reviews. Jan 2018;84:218224. doi:10.1016/j.neubiorev.2017.11.021 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold iew ed 64. Spring Harb Perspect Biol. Sep 4 2014;6(10):a016295. doi:10.1101/cshperspect.a016295 65. Recasens M, Almolda B, Pérez-Clausell J, Campbell IL, González B, Castellano B. Chronic exposure to IL-6 induces a desensitized phenotype of the microglia. Journal of neuroinflammation. Jan 22 2021;18(1):31. doi:10.1186/s12974-020-02063-1 Yao X, Huang J, Zhong H, et al. Targeting interleukin-6 in inflammatory autoimmune ev 66. diseases and cancers. Pharmacol Ther. Feb 2014;141(2):125-39. 67. er r doi:10.1016/j.pharmthera.2013.09.004 Liu Y, Ho RC, Mak A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a doi:10.1016/j.jad.2011.08.003 68. pe meta-analysis and meta-regression. Journal of affective disorders. Aug 2012;139(3):230-9. Lyra e Silva NM, Gonçalves RA, Pascoal TA, et al. Pro-inflammatory interleukin-6 ot signaling links cognitive impairments and peripheral metabolic alterations in Alzheimer’s disease. Translational Psychiatry. 2021/04/28 2021;11(1):251. doi:10.1038/s41398-021-01349-z Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-Brain Barrier Dysfunction tn 69. Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain rin Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Review. Frontiers in cellular neuroscience. 2021-September-13 2021;15doi:10.3389/fncel.2021.661838 Yang J, Ran M, Li H, et al. New insight into neurological degeneration: Inflammatory ep 70. cytokines and blood–brain barrier. Review. Frontiers in Molecular Neuroscience. 2022-October- Pr 24 2022;15doi:10.3389/fnmol.2022.1013933 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Wajant H, Siegmund D. TNFR1 and TNFR2 in the Control of the Life and Death iew ed 71. Balance of Macrophages. Review. Frontiers in Cell and Developmental Biology. 2019-May-29 2019;7doi:10.3389/fcell.2019.00091 72. Cock HR, Edwards MJ. Functional neurological disorders: acute presentations and management. Clin Med (Lond). Oct 2018;18(5):414-417. doi:10.7861/clinmedicine.18-5-414 Valerio MP, Lomastro J, Igoa A, Martino DJ. Clinical Characteristics of Melancholic and ev 73. Nonmelancholic Depressions. J Nerv Ment Dis. Mar 1 2023;211(3):248-252. 74. er r doi:10.1097/nmd.0000000000001616 Sumner JA, Hagan K, Grodstein F, Roberts AL, Harel B, Koenen KC. Posttraumatic stress disorder symptoms and cognitive function in a large cohort of middle-aged women. doi:https://doi.org/10.1002/da.22600 75. pe https://doi.org/10.1002/da.22600. Depression and Anxiety. 2017/04/01 2017;34(4):356-366. Franklin AR, Mathersul DC, Raine A, Ruscio AM. Restlessness in Generalized Anxiety ot Disorder: Using Actigraphy to Measure Physiological Reactions to Threat. Behav Ther. May 2021;52(3):734-744. doi:10.1016/j.beth.2020.09.004 Association AP. Diagnostic and statistical manual of mental disorders. Fifth ed. 2013. 77. Kamiya K, Hori M, Aoki S. NODDI in clinical research. Journal of Neuroscience tn 76. 78. rin Methods. 2020/12/01/ 2020;346:108908. doi:https://doi.org/10.1016/j.jneumeth.2020.108908 Taylor WD, Kudra K, Zhao Z, Steffens DC, MacFall JR. Cingulum bundle white matter ep lesions influence antidepressant response in late-life depression: a pilot study. Journal of affective disorders. Jun 2014;162:8-11. doi:10.1016/j.jad.2014.03.031 Mertse N, Denier N, Walther S, et al. Associations between anterior cingulate thickness, Pr 79. cingulum bundle microstructure, melancholia and depression severity in unipolar depression. This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
doi:https://doi.org/10.1016/j.jad.2022.01.035 80. iew ed Journal of affective disorders. 2022/03/15/ 2022;301:437-444. Kim SJ, Jeong DU, Sim ME, et al. Asymmetrically altered integrity of cingulum bundle in posttraumatic stress disorder. Neuropsychobiology. 2006;54(2):120-5. doi:10.1159/000098262 81. Weis CN, Belleau EL, Pedersen WS, Miskovich TA, Larson CL. Structural Connectivity ev of the Posterior Cingulum Is Related to Reexperiencing Symptoms in Posttraumatic Stress Disorder. Chronic Stress (Thousand Oaks). Jan-Dec 2018;2doi:10.1177/2470547018807134 Espay AJ, Ries S, Maloney T, et al. Clinical and neural responses to cognitive behavioral er r 82. therapy for functional tremor. Neurology. Nov 5 2019;93(19):e1787-e1798. doi:10.1212/wnl.0000000000008442 Bubb EJ, Metzler-Baddeley C, Aggleton JP. The cingulum bundle: Anatomy, function, pe 83. and dysfunction. Neuroscience & Biobehavioral Reviews. 2018/09/01/ 2018;92:104-127. doi:https://doi.org/10.1016/j.neubiorev.2018.05.008 Choi KW, Jang EH, Kim AY, et al. Predictive inflammatory biomarkers for change in ot 84. suicidal ideation in major depressive disorder and panic disorder: A 12-week follow-up study. J 85. tn Psychiatr Res. Jan 2021;133:73-81. doi:10.1016/j.jpsychires.2020.12.011 Köhler CA, Freitas TH, Maes M, et al. Peripheral cytokine and chemokine alterations in rin depression: a meta-analysis of 82 studies. Acta Psychiatr Scand. May 2017;135(5):373-387. doi:10.1111/acps.12698 Ferencova N, Visnovcova Z, Ondrejka I, et al. Evaluation of Inflammatory Response ep 86. System (IRS) and Compensatory Immune Response System (CIRS) in Adolescent Major Pr Depression. J Inflamm Res. 2022;15:5959-5976. doi:10.2147/jir.S387588 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791
Gao W, Xu Y, Liang J, et al. Comparison of serum cytokines levels in normal-weight and iew ed 87. overweight patients with first-episode drug-naïve major depressive disorder. Front Endocrinol (Lausanne). 2022;13:1048337. doi:10.3389/fendo.2022.1048337 88. Burrows K, Stewart JL, Kuplicki R, et al. Elevated peripheral inflammation is associated with attenuated striatal reward anticipation in major depressive disorder. Brain Behav Immun. 89. ev Mar 2021;93:214-225. doi:10.1016/j.bbi.2021.01.016 Black DS, Slavich GM. Mindfulness meditation and the immune system: a systematic 2016;1373(1):13-24. doi:10.1111/nyas.12998 90. er r review of randomized controlled trials. Annals of the New York Academy of Sciences. Jun Creswell JD, Taren AA, Lindsay EK, et al. Alterations in Resting-State Functional pe Connectivity Link Mindfulness Meditation With Reduced Interleukin-6: A Randomized Controlled Trial. Biol Psychiatry. Jul 1 2016;80(1):53-61. doi:10.1016/j.biopsych.2016.01.008 91. Hoge EA, Bui E, Palitz SA, et al. The effect of mindfulness meditation training on ot biological acute stress responses in generalized anxiety disorder. Psychiatry Res. Apr 2018;262:328-332. doi:10.1016/j.psychres.2017.01.006 Litton CE. A clinician's perspective in the management of functional seizures. Epilepsy tn 92. Pr ep rin Behav Rep. 2021;16:100492. doi:10.1016/j.ebr.2021.100492 This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=4435791