PreprintPDF Available

Longitudinal Alterations of the Cisternal Segment of Trigeminal Nerve and Brain Pain-matrix Regions in Patients with Trigeminal Neuralgia Before and After Treatment

Authors:

Abstract

Background: Trigeminal neuralgia (TN) is the most common type of chronic neuropathic facial pain, but the etiology and pathophysiological mechanisms after treatment are still not well understood. The purpose of this study was to investigate the longitudinal changes of the cisternal segment of the trigeminal nerve and brain pain-related regions in patients with TN before and after treatment using readout segmentation of long variable echo-train (RESOLVE) diffusion tensor imaging (DTI) and transverse relaxation (T2)-weighted sampling perfection with application-optimized contrast at different flip angle evolutions (T2-SPACE). Methods: Twelve patients with TN and four healthy controls were enrolled in this study. All patients underwent assessment of the visual analog scale (VAS), and acquisition of RESOLVE DTI and T2-SPACE images before and at 1, 6, and 12 months after treatments. Regions-of-interest were placed on the bilateral anterior, middle, and posterior parts of the cisternal segment of the trigeminal nerve, the bilateral root entry zone (REZ), bilateral nuclear zone, and the center of pontocerebellar tracts, respectively. Voxel-based morphometry (VBM) analysis was conducted with T2-SPACE images, and gray matter volumes (GMV) were measured from brain pain-matrix regions. Results: The results demonstrated that the axial diffusivity of the middle part of the cisternal trigeminal nerve, the fractional anisotropy of the bilateral nuclear zones, and the mean diffusivity of the center of pontocerebellar tract significantly changed over time before and after treatment. The changes of GMV in the pain-matrix regions exhibited similar trends to the VAS before and after treatment. Conclusion: We conclude that magnetic resonance imaging with RESOLVE DTI and VBM with T2-SPACE images were helpful in the understanding of the pathophysiological mechanisms in patients with TN before and after treatment.
Page 1/15
Longitudinal Alterations of the Cisternal Segment of
Trigeminal Nerve and Brain Pain-matrix Regions in
Patients with Trigeminal Neuralgia Before and After
Treatment
Tai-Yuan Chen
Chi-Mei Medical Center
Ching-Chung Ko
Chi-Mei Medical Center
Te-Chang Wu
Chi-Mei Medical Center
Li-Ching Lin
Chi-Mei Medical Center
Yun-Ju Shih
Chi-Mei Medical Center
Yi-Chieh Hung
Chi-Mei Medical Center
Ming-Chung Chou ( mcchou@kmu.edu.tw )
Kaohsiung Medical University
Research Article
Keywords: Trigeminal Neuralgia, Trigeminal Nerve, Pain-matrix regions, RESOLVE DTI, T2-SPACE VBM
DOI: https://doi.org/10.21203/rs.3.rs-289655/v1
License: This work is licensed under a Creative Commons Attribution 4.0 International License. 
Read Full License
Page 2/15
Abstract
Background: Trigeminal neuralgia (TN) is the most common type of chronic neuropathic facial pain, but
the etiology and pathophysiological mechanisms after treatment are still not well understood. The
purpose of this study was to investigate the longitudinal changes of the cisternal segment of the
trigeminal nerve and brain pain-related regions in patients with TN before and after treatment using
readout segmentation of long variable echo-train (RESOLVE) diffusion tensor imaging (DTI) and
transverse relaxation (T2)-weighted sampling perfection with application-optimized contrast at different
ip angle evolutions (T2-SPACE).
Methods: Twelve patients with TN and four healthy controls were enrolled in this study. All patients
underwent assessment of the visual analog scale (VAS), and acquisition of RESOLVE DTI and T2-SPACE
images before and at 1, 6, and 12months after treatments. Regions-of-interest were placed on the
bilateral anterior, middle, and posterior parts of the cisternal segment of the trigeminal nerve, the bilateral
root entry zone (REZ), bilateral nuclear zone, and the center of pontocerebellar tracts, respectively. Voxel-
based morphometry (VBM) analysis was conducted with T2-SPACE images, and gray matter volumes
(GMV) were measured from brain pain-matrix regions.
Results: The results demonstrated that the axial diffusivity of the middle part of the cisternal trigeminal
nerve, the fractional anisotropy of the bilateral nuclear zones, and the mean diffusivity of the center of
pontocerebellar tract signicantly changed over time before and after treatment. The changes of GMV in
the pain-matrix regions exhibited similar trends to the VAS before and after treatment.
Conclusion: We conclude that magnetic resonance imaging with RESOLVE DTI and VBM with T2-SPACE
images were helpful in the understanding of the pathophysiological mechanisms in patients with TN
before and after treatment.
Introduction
Trigeminal neuralgia (TN) is the most common type of chronic neuropathic facial pain [1] and is
characterized by intermittent attacks of severe, electric shock-like unilateral pain along the distribution of
the trigeminal nerve branches [2]. Although not essential to TN pathophysiology, a well-established
etiological factor is the neurovascular compression (NVC) of the trigeminal nerve at the root entry zone
(REZ) [3–5], where focal demyelination is believed to occur at the point of contact [6, 7]. The surgical
treatments performed, including Gamma/Cyber Knife radiosurgery (CKRS), microvascular decompression
surgery, and percutaneous trigeminal rhizotomy, have been previously described [8, 9].
Advances in neuroimaging techniques have identied key neuroanatomical signatures. It is well known
that magnetic resonance imaging (MRI) with diffusion tensor imaging (DTI) is capable of depicting
microstructural changes of the trigeminal nerve [10]. With the application of readout segmentation of
long variable echo-train (RESOLVE) DTI and parallel imaging [11, 12], susceptibility distortions could be
Page 3/15
reduced substantially to analyze more accurately subtle changes involving the symptomatic cisternal
segment of trigeminal nerve, REZ, and the nuclear zone.
In addition, high-resolution longitudinal relaxation (T1)-weighted images were previously utilized to detect
whole-brain gray matter (GM) changes using voxel-based morphometric (VBM) analysis. Given that the
evaluation of the brain's GM could provide insights into disease mechanisms, it is helpful to use VBM
analysis in the detection of GM abnormalities in patients with neuralgia. Previously, brain GM
abnormalities have been identied in patients with chronic pain [13]. The most common locations of GM
abnormalities include regions implicated in the multidimensional experience of pain, such as the
prefrontal cortex (PFC), insula, anterior cingulate cortex (ACC) and mid-cingulate cortex), thalamus,
primary and secondary somatosensory cortices (S1, S2), basal ganglia, amygdala, and brainstem [13].
These abnormalities are commonly found for many types of chronic pain, including those that affect the
trigeminal system, such as migraines [14], trigeminal neuropathic pain [15, 16], and temporomandibular
disorders [17]. Given that some abnormalities are common across most chronic pains (e.g., cortical
thinning in the anterior insula, cingulate cortex, and dorsolateral PFC) [13], the changes of these areas
likely reect pain chronicity in general and may be associated with negative effect and changes in pain
modulation [18]. In a previous TN study, the reductions of the volumes of the ACC and the superior
temporal gyrus (STG)/middle temporal gyrus were documented [19]. It was also considered that the GM
volume (GMV) of the STG decreased as the duration of disease increased. Such MR-detectable brain
structural alterations may reect changes in the neuronal size or number, synaptogenesis, dendritic
branching, axon sprouting, synaptic pruning, neuronal cell death, alterations in vasculature, and the sizes
or numbers of glial cells [20, 21].
Moreover, transverse relaxation (T2)-weighted sampling with application-optimized contrast with different
ip angle evolution (T2-SPACE) images were used to evaluate cortical/subcortical brain GM based on
VBM analyses [22]. However, T2-SPACE images have not been previously utilized to investigate
longitudinal brain changes in TN subjects after surgeries. As the surgical treatments could relieve pain
symptoms in patients, it is of importance to understand how the cisternal segment of the trigeminal nerve
and brain GM structures were changed after the treatments. Therefore, the purposes of this study were to
evaluate longitudinal diffusion changes of the cisternal segment of trigeminal nerves using RESOLVE DTI
technique and to assess longitudinal brain GM differences using T2-SPACE VBM analysis in TN patients
after CKRS or radiofrequency ablation (RFA).
Materials And Methods
Patients
The study received institutional approval from the Human Research Ethics Committee of our hospital
(Institutional Review Board 10603-003), and written informed consents were obtained from all
participants.
Page 4/15
We enrolled 12 patients with refractory TN that lasted for more than 2 years, who received CKRS or RFA
treatments from January 2016 to December 2018. In addition, four age- and sex-matched healthy
controls (2M/2F, age = 63.3 ± 4.4 years) were enrolled for comparisons.
Data Acquisition
After providing informed consent, all subjects underwent pain intensity assessments of the pain intensity
and the severity of neuralgia using the visual analog scale (VAS). MRI scans were also conducted to
exclude the possibility of occupying lesions. In this study, MRI was conducted on a 1.5-T scanner
equipped with a 24-channel phased-array head coil. The standard MRI protocol included the following:
axial T1-weighted imaging, T2-weighted imaging, uid attenuated inversion recovery, T2-weighted
gradient-recalled echo imaging, MR angiography, and single-shot spin-echo echo-planar diffusion-
weighted imaging. In addition, during the same imaging session and follow-up sessions at 1, 6, and 12
months after treatment, all subjects underwent three-dimensional (3D), whole-brain, high-resolution T2-
SPACE imaging (TR/TE = 2200/276 ms, echo train length = 150, number of slice = 192, matrix size = 
256×256, voxel size = 1×1×1 mm3), in which an imaging plane parallel to cisternal segments of trigeminal
nerves was placed to acquire both single-shot DTI (TR/TE = 6000/87 ms, number of slice = 21, slice
thickness = 2 mm, no gap, FOV = 22 cm, matrix size = 128×128, number of excitation = 1, acceleration
factor = 2, b-values = 0 and 1000 s/mm2) and RESOLVE DTI (TR/TE = 3330/75 ms, number of slice = 21,
slice thickness = 2 mm, no gap, FOV = 22 cm, matrix size = 128×128, number of excitation = 1, number of
segment = 4, acceleration factor = 2, b-values = 0 and 1000 s/mm2) with 30 diffusion directions.
Image Processing and Analysis
All imaging data were transferred to a stand-alone workstation and post-processed with FSL (FMRIB
Software Library, Oxford, UK) and MATLAB (MathWorks Inc., Natick, MA, USA). First, the DTI data were
motion-corrected using rigid-body registration, and the diffusion directions were compensated by the
rotation matrix. Second, eddy-current distortions were minimized using 12-parameter ane image
registration. Third, the fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity (RD), and mean
diffusivity (MD) values in each voxel were calculated with the DTIFIT tool. Fourth, the DTI metrics were
analyzed in the trigeminal nerve cisternal segment on the affected and contralateral sides based on the
selection of a region-of-interest (ROI) with custom-made software that ran on a MATLAB platform that
automatically divided the trigeminal nerve cisternal segment into anterior, middle, and posterior parts, as
shown in Fig.1.
Moreover, individual FA maps were spatially transformed to a representative FA map—which used the
minimal transformation distance among all patients—using linear ane and nonlinear demon
registrations. Subsequently, ROIs were placed on the REZ, nuclear zone, and the center of pontocerebellar
tracts (PCT) in the pons, as shown in Fig.1. Finally, the mean FA, AD, RD, and MD values in these regions
were calculated for statistical analysis.
Page 5/15
The whole-brain T2-SPACE images were used in VBM analysis to estimate the brain GMV with the
Computational Anatomical Toolbox version 12, (CAT12) and Statistical Parametric Mapping version 12
(SPM12) in MATLAB. First, T2-SPACE images were segmented into GM, white matter, and cerebrospinal
uid, spatially normalized with diffeomorphic anatomical registration through exponentiated Lie algebra
(DARTEL) to the subject-specic template. The segmented images were then smoothed with an isotropic
Gaussian kernel with a full-width-at-half-maximum equal to 8 mm. Subsequently, an automated
anatomical labeling brain template was used to separate the brain cortex into 116 regions, and mean
GMVs were calculated in the pain-related regions, including the periaqueductal GM (PAG), PFC, posterior
cingulate cortex (PCC), ACC, insula, amygdala, thalamus, posterior parietal cortex (PPC), S1, S2,
supplementary motor area (SMA), and cerebellum [23].
Statistical Analysis
A Wilcoxon signed-rank test was performed to understand the difference of DTI indices in the cisternal
segments of the trigeminal nerve, REZ, nuclear zone, and the center of PCT, and the difference of GMV in
pain-related brain regions in healthy controls and patients between the two hemispheres, respectively. The
results were considered signicant if P < 0.05.
In addition, one-way analysis of variation (ANOVA) was performed to show whether the DTI indices
changed along the three parts of the cisternal segments of the trigeminal nerve and whether the DTI
indices of the trigeminal nerve, REZ, nuclear zone, center of PCT, the GMV of pain-related brain regions,
and the VAS, signicantly changed over time before and after treatment, respectively. The results were
considered signicant if P < 0.05.
Moreover, Pearson's correlational analysis was performed to reveal the relationship between DTI indices
of trigeminal nerve, REZ, nuclear zone, the center of PCT, and the GMV of pain-related brain regions. The
correlations were considered signicant if P < 0.005.
Results
The mean age of the 12 enrolled patients was 68.9 years, including four men and eight women. All of
them had TN unilaterally, with 11 on the right side and 1 on the left side. None of them had denite space-
occupying lesions. All patients successfully underwent VAS assessments before and at 1, 6, and 12
months after treatment. However, in the imaging study, although all patients underwent MRI prior to the
treatment, six patients did not undergo a follow-up MRI at 1 and 6 months, and eight patients did not
undergo a follow-up MRI at 12 months after treatment owing to refusals or dropout. The demographic
and clinical characteristics of the enrolled patients are listed in Table1.
Page 6/15
Table 1
Demographic characteristics of patients with trigeminal neuralgia (TN) enrolled in this study.
Patient NVC VAS score Procedure
Before Tx 1
month 6 months 12 months
1 NO 7 0 0 0 CKRS
2 Rt. REZ 8 0 0 0 CKRS
3 Lt. REZ 9 1 0 0 RFA
4 Rt. REZ 9 0 9 2 RFA, CKRS
5 Rt. REZ 10 0 0 0 RFA, CKRS
6 Rt. REZ 9 0 0 0 RFA
7 Rt. REZ 9 0 2 4 CKRS, RFA
8 NO 10 0 0 0 CKRS
9 Rt. REZ 10 0 0 0 CKRS
10 Rt. REZ 10 0 0 0 RFA
11 Lt. middle part 10 0 0 0 RFA
12 NO 10 0 0 0 CKRS
Abbreviations: NVC = neurovascular compression, Tx = treatment, REZ = root entry zone, VAS = visual
analog scale, CKRS = cyber knife radiosurgery, RFA = radiofrequency ablation.
 The diagnostic MRI demonstrated that among the 12 patients, eight had NVC on the trigeminal nerve
REZ, one had NVC at the middle part of the cisternal segment of trigeminal nerve, and three had no NVC
signs. In addition, six patients received RFA, and others received CKRS treatment. However, after
treatment, two patients experienced TN relapse at 6 months and received an additional treatment using
CKRS or RFA. In the VAS analysis, the results showed that the initial VAS was as high as 9.25 ± 0.97 (10
was the highest score). After 1 month of treatment, the VAS was signicantly reduced to 0.08 ± 0.29.
However, the VAS was slightly increased to 0.92 ± 2.61 at 6 months but was decreased to 0.50 ± 1.24 at
12 months after treatment. The ANOVA analysis revealed that the VAS signicantly changed over time
before and after treatment.
Diffusion changes using RESOLVE DTI technique
In patients with TN, the entire cisternal segment of the trigeminal nerve exhibited no signicant difference
in DTI indices between the affected and contralateral sides before and at 1, 6, and 12 months after
treatment. In the anterior part of the cisternal trigeminal nerve, no signicant difference of FA, AD, RD, and
MD values was noted between the two sides before treatment, but signicant AD differences were noted
Page 7/15
between the two sides at 1 month after treatment. Additionally, AD was signicantly increased from 1 to 6
months after treatment in the affected side (Fig.2). However, in healthy controls, no signicant
differences in the DTI indices were noted between the two sides.
In the middle part of the cisternal trigeminal nerve, the results yielded no signicant differences for FA, AD,
RD, and MD values between the two sides before, and at 1, 6, and 12 months after treatment, whereas RD
and MD values were signicantly decreased from 1 to 6 months after treatment in the affected side in
patients with TN, as shown in Fig.3. However, in healthy controls, no signicant differences were noted in
the DTI indices between the two sides.
In the posterior part of the cisternal trigeminal nerve, no signicant difference was noted in FA, AD, RD,
and MD values between the two sides in patients with TN before and at 1, 6, and 12 months after
treatment. However, the FA value was signicantly increased from 1 to 6 months after treatment in the
affected side, as shown in Fig.4. In healthy controls, no signicant differences were noted in the DTI
indices between the two sides. The ANOVA analysis further revealed that only the AD values signicantly
changed over time (P = 0.034) in the middle part of the cisternal trigeminal nerve of the affected side
before and after treatment.
In the REZ, the results yielded signicant differences for the FA values between the two sides in the
patients with TN before and 6 months after treatment and signicant differences for the AD, RD, and MD
values before and at 1 and 6 months after treatment. In the affected side, the FA was signicantly
increased 1 month after treatment but was signicantly decreased from 1 to 6 months after treatment. In
the contralateral side, however, the FA value was signicantly reduced 1 month after treatment, as shown
in Fig.5. In healthy controls, no signicant differences in the DTI indices were noted between the two
sides. The ANOVA analysis showed no signicant changes in the DTI indices over time in both sides
before and after treatment.
In the nuclear zone, the results yielded signicant differences in the cases of the FA, AD, and MD values
between the two sides in patients with TN before treatment and signicant FA value differences 6 months
after treatment. However, in both sides, the FA value was signicantly reduced 1 month after treatment
but was signicantly increased from 1 to 6 months after treatment, as shown in Fig.6. In healthy
controls, no signicant differences in the DTI indices were noted between the two sides. Additionally, the
ANOVA analysis revealed that the FA value signicantly changed over time in the nuclear zones of both
sides (P = 0.0002 in the normal side, P = 0.0389 in the lesion side) before and after treatment.
In the center of PCT, the FA and AD values were slightly decreased, whereas the RD and MD values were
slightly increased in patients with TN 1 month after treatment. No signicant difference was noted
between two time points, as shown in Fig.7. However, the ANOVA analysis revealed that the MD value
signicantly increased over time (P = 0.00069) in the center of PCT before and after treatment.
T2-SPACE VBM analysis
Page 8/15
In the VBM analysis, the results revealed that GMV was signicantly lower in the affected side compared
with the contralateral side in the PCC, ACC, insula, amygdala, and S1 in the patients before treatment.
After 1 month of treatment, no signicant GMV difference was noted in the pain-related regions between
the two hemispheres. However, the GMV was signicantly lower in the affected side compared with the
contralateral side in the PCC, insula, amygdala, and S2 at 6 months after treatment, but the difference no
longer existed in the pain-related regions between the two hemispheres at 12 months after treatment, as
shown in Fig.8. In healthy controls, no signicant differences in the GMVs were noted between the two
hemispheres. The ANOVA analysis showed that the GMVs did not signicantly change over time in the
pain-matrix regions of the affected and contralateral sides before and after treatment.
In correlational analysis, the results revealed that before treatment, the FA of the anterior part of the
cisternal trigeminal nerve in the affected side was signicantly correlated with the GMV of PAG, bilateral
PFC, bilateral amygdala, bilateral SMA, bilateral cerebellum, left S1, left PCC, left insula, right PPC, and
right ACC. Moreover, the AD of the middle part of the cisternal trigeminal nerve in the affected side was
signicantly correlated with the GMV of the bilateral PFC, bilateral PPC, and left S2, and the RD and MD
values of the middle part of the cisternal trigeminal nerve in the affected side were also signicantly
correlated with the GMV of the right S2, as shown in Table2.
Page 9/15
Table 2
Signicant correlation coecients between DTI indices of cisternal trigeminal nerve and gray matter
volume (GMV) of pain-matrix regions in patients with TN before treatment.
Region FA (anterior portion
of the affected side) AD (middle part of
the affected side) RD (middle part of
the affected side) MD (middle part of
the affected side)
PAG 0.8319 -- -- --
Lt. PFC 0.8454 0.7779 -- --
Rt. PFC 0.8632 0.7916 -- --
Lt. PCC 0.7500 -- -- --
Rt. ACC 0.7560 -- -- --
Lt.
amygdala 0.7807 -- -- --
Rt.
amygdala 0.8418 -- -- --
Lt. PPC -- 0.8114 -- --
Rt. PPC 0.7889 0.7818 -- --
Lt. SMA 0.8503 -- -- --
Rt. SMA 0.8859 -- -- --
Lt. S1 0.7611 -- -- --
Lt. S2 -- 0.8397 -- --
Rt. S2 -- -- 0.7658 0.8137
Lt.
cerebellum 0.8215 -- -- --
Rt.
cerebellum 0.7898 -- -- --
Abbreviations: FA = fractional anisotropy, AD = axial diffusivity, RD = radial diffusivity, MD = mean
diffusivity, PAG = periaqueductal gray, PFC = prefrontal cortex, PCC = posterior cingulate cortex, ACC = 
anterior cingulate cortex, SMA = supplementary motor area, S1 = primary somatosensory cortex, S2 = 
secondary somatosensory cortex.
However, after 1 month of treatment, only the FA of the middle part of the cisternal trigeminal nerve in the
affected side yielded a signicant correlation with the GMV of PAG (cc = 0.9759). After 6 months of
treatment, only the RD of the nuclear zone in the affected side yielded a signicant correlation with the
GMV in the right S1 (cc = 0.9537). After 12 months of treatment, only the MD of the nuclear zone in the
affected side yielded a signicant correlation with the GMV in the left thalamus (cc = 0.9957).
Discussion
Page 10/15
To the best of our knowledge, this is the rst study that conducted both RESOLVE DTI and 3D T2-SPACE
imaging to investigate the longitudinal changes of the cisternal segment of the trigeminal nerve, REZ,
nuclear zone, the center of PCT, and brain pain-matrix regions in the patients with TN before and after
treatment. Several ndings presented herein may help understand the etiology and pathophysiology of
TN. First, the trigeminal nerve REZ and nuclear zone exhibited more changes than the cisternal segment
of the trigeminal nerve and the GMVs of brain pain-matrix regions before and after treatment. Second, the
contralateral REZ and nuclear zone were signicantly altered after treatment most likely owing to
Wallerian degeneration. Third, the AD of the middle part of the cisternal trigeminal nerve, the FA of
bilateral nuclear zones, and the MD of the center of PCT signicantly changed over time before and after
treatment. Fourth, the difference of GMV in the pain-matrix regions between the two hemispheres
exhibited similar trends to the VAS before and after treatment. Finally, the DTI indices in the cisternal
trigeminal nerve (FA) and nuclear zone (RD and MD) of the affected side were signicantly correlated
with the GMVs in specic pain-matrix regions (PAG, right S1, and left thalamus) before and after
treatment. These ndings are further discussed in the following parts of the document.
In the DTI analysis, the results showed that patients with TN exhibited slightly lower AD and FA values
with no statistical signicance in the affected side of the cisternal segment of the trigeminal nerve before
treatment. The signicant changes in the AD, RD, MD, and FA values in the affected side after treatment
suggested that the treatments signicantly altered the microstructural diffusion in the trigeminal nerve. In
contrast, the REZ and nuclear zones yielded signicant differences in the values of the DTI indices
between the two sides before and after treatment, thus indicating that TN led to more tissue alterations in
the REZ and nuclear zones than the cisternal trigeminal nerve, most likely owing to the fact that 67%
(8/12) of studied patients exhibited NVC at REZ.
Our study also demonstrated that patients with TN not only had lower FA but also had higher AD, RD, and
MD values at the affected REZ. Consistent with a prior study [24], our ndings suggested that the
increased MD and RD may be linked to NVC-induced focal demyelination in the trigeminal nerve REZ [25],
neuroinammatory processes, and/or edema [26] that affected the trigeminal system. Therefore, DTI can
detect subtle pathological features at the trigeminal nerve REZ supporting a role for this regional
involvement in TN pathophysiology. Moreover, the present study performed the treatments with a focus
on the middle part of the cisternal trigeminal nerve in the affected side. Thus, the signicant changes of
the FA values in the distal and contralateral REZ and nuclear zone suggested that the Wallerian
degeneration occurred through the PCT [27, 28], wherein minor changes of the DTI indices were observed
after treatment.
The present study employed 3D T2-SPACE images to examine cortical/subcortical brain GMVs based on
VBM analysis. A previous study reported that patients with TN had greater GMVs in the thalamus,
contralateral S1, amygdala, frontal pole, PAG, primary motor cortex, and basal ganglia and cortical
thinning in the orbitofrontal cortex, pregenual ACC, and insula [29], whereas anther study reported that
patients with TN showed GMV reductions including the frontal, temporal, and parietal areas, as well as in
the left thalamus and right cerebellum [30]. Our study found that patients with TN had signicantly lower
Page 11/15
GMVs in PCC, ACC, insula, amygdala, and S1 in the affected side compared with the contralateral side
before treatment. These ndings may reect unique symptoms because TN is characterized by
paroxysmal pain triggered by innocuous stimuli or movements and does not involve major sensory
losses. However, the differences between the previous ndings and ours may be attributed to the mixed
trigeminal pain patient groups and the different methodology (T1-VBM versus T2-VBM) used to assess
GM changes.
In addition, our results demonstrated that TN led to signicant GMV differences between the two
hemispheres in some pain-related brain regions (PCC, ACC, insula, amygdala, and S1) and that the
therapy helped reduce the GMV difference at 1 month after treatment. However, the GMV differences in
some pain-related regions (PCC, insula, amygdala, and S2) became signicant again at 6 months after
treatment but returned to insignicant outcomes at 12 months after treatment. The sequential changes of
GMV in the pain-matrix regions were generally consistent with the VAS in patients with TN. Initially, the
pain intensity was measured to e as high as 9.25 ± 0.97 before treatment and was signicantly reduced
to 0.08 ± 0.29 after 1 month of treatment. However, at 6 months after treatment, two patients who
exhibited degraded pain intensity, which resulted in a slight increase in VAS (0.92 ± 2.61), received a
second treatment. After the second treatment, one patient had improved symptoms with the VAS reduced
from 9 to 2, but the other had deteriorated symptoms with a VAS increased from 2 to 4. As a result, the
overall VAS was slightly decreased after 12 months of treatment. Moreover, the correlation analysis
revealed signicant correlations between the DTI indices of the cisternal trigeminal nerve and nuclear
zone and the GMVs of the pain-matrix regions in the affected side before and after treatment. These
ndings suggested that the changes of GMV in the pain-matrix regions were likely associated with the
changes of VAS and that VBM analysis of T2-SPACE was helpful in the noninvasive monitoring and
reection of the pain intensity in patients with TN before and after treatment.
The etiology and pathophysiological mechanisms of TN are still not well understood, and the central
contributions in TN are still debated. The most common theory of the classical TN etiology is a peripheral
theory that involves the compression of the trigeminal nerve's REZ by blood vessels owing to the surgical
experience [5]. Moreover, several studies demonstrated pathological changes and most notably the
demyelination of the trigeminal nerves in patients with TN [6, 31–35]. In the present study, despite the
fact that minor changes were observed in the DTI indices in the cisternal segment of the trigeminal nerve,
no statistical signicance was revealed between the affected and contralateral sides before treatment.
Nevertheless, the REZ and nuclear zone yielded signicant changes in the DTI indices before treatment
and more signicant changes than the cisternal segment of trigeminal nerve after treatment. This
suggested that the REZ and nuclear zone were more sensitive to TN before treatment and had more
demyelination after treatment compared with the cisternal segment of the trigeminal nerve.
However, the NVC theory of TN cannot suciently explain the disorder because some individuals can
develop TN in the absence of NVC, and some individuals with NVC never develop TN. In the present study,
25% (3/12) of the studied patients developed TN without NVC. This suggests that the TN symptoms were
not fully attributable to the presence of NVC. It is known that peripheral nerve injury can lead to central
Page 12/15
nervous system (CNS) plasticity [36, 37] most likely owing to the sustained or repetitive activation of
primary afferent bers and central sensitization [38]. In the present study, our results demonstrated that
the changes of DTI indices in the trigeminal nerve were associated with the alterations of GMVs in brain
pain-matrix regions before and after treatment. Furthermore, the longitudinal changes of GMV in the pain-
matrix regions were generally consistent with the sequential changes of VAS, thus suggesting that the
CNS changes in conjunction with trigeminal nerve injury contribute to TN symptoms.
The study is associated with some limitations. First, the small sample size of this study may lead to a
low statistical power. Thus, the results ought to be interpreted with care. Second, some patients did not
undergo a follow-up MRI scan owing to refusal or dropout. Thus, the incomplete dataset may affect the
statistical results. Third, comparisons were not conducted between patients with TN and healthy controls
in DTI and VBM analysis because only four age- and sex-matched healthy controls were enrolled in this
study. Further investigations with larger groups will be needed to comprehensively compare the
differences of cisternal trigeminal nerve and brain pain-matrix regions among healthy subjects and
patients with TN before and after treatment.
Conclusions
MRI with RESOLVE DTI was capable of detecting microstructural changes in patients with refractory TN
before and after treatment, as demonstrated by the longitudinal changes of the FA, AD, RD, and MD
values on the symptomatic side of the trigeminal nerve cisternal segment, REZ, nuclear zone, and PCT.
The VBM analysis of T2-SPACE data was helpful in detecting longitudinal changes of GMV in the pain-
matrix regions on the symptomatic cerebral hemisphere in the studied patients before and after
treatment. In this study, we discussed theories of TN pathophysiology, assessed how advanced
neuroimaging methods contribute to our current understanding of TN, and outlined future directions for
clinical trials that may use these methods to allow treatment selection and response for patients with TN
based on brain and/or nerve biomarkers.
Declarations
Ethics approval and consent to participate
The study was approved by local institution review board (10603-003) of Chi Mei Medical Center.
Consent for publication
Inform consent was obtained from all subjects. The study was carried out in accordance with Helsinki
Declaration.
Availability of data and materials
Page 13/15
The data presented in this study are available on request from the corresponding author. The data are not
publicly available due to the nature of this research, participants of this study did not agree for their data
to be shared publicly.
Competing interests
No conict of interest exists in this study.
Funding
This study was funded by Chi-Mei Medical Center (CMFHR10617).
Authors' contributions
Chen TY, Lin CL, Hung YC, and Chou MC designed the study. Chen TY, Ko CC, Wu TC, and Shih YJ
collected the patient data. Lin LC and Hung YC performed the treatment. Chen TY and Chou MC analyzed
and wrote the original draft. All authors reviewed and edited the manuscript.
Acknowledgements
This work was supported by a grant from the Chi-Mei Medical Center (CMFHR10617).
References
1. Koopman JSHA, Dieleman JP, Huygen FJ, de Mos M, Martin CGM, Sturkenboom MCJM: Incidence of
facial pain in the general population.
Pain
2009, 147(1-3):122-127.
2. Eller JL, Raslan AM, Burchiel KJ: Trigeminal neuralgia: denition and classication.
Neurosurg Focus
2005, 18(5):E3.
3. Antonini G, Di Pasquale A, Cruccu G, Truini A, Morino S, Saltelli G, Romano A, Trasimeni G, Vanacore
N, Bozzao A: Magnetic resonance imaging contribution for diagnosing symptomatic neurovascular
contact in classical trigeminal neuralgia: a blinded case-control study and meta-analysis.
Pain
2014,
155(8):1464-1471.
4. Maarbjerg S, Wolfram F, Gozalov A, Olesen J, Bendtsen L: Signicance of neurovascular contact in
classical trigeminal neuralgia.
Brain
2015, 138(Pt 2):311-319.
5. Nurmikko TJ, Eldridge PR: Trigeminal neuralgia--pathophysiology, diagnosis and current treatment.
Br
J Anaesth
2001, 87(1):117-132.
6. Devor M, Govrin-Lippmann R, Rappaport ZH: Mechanism of trigeminal neuralgia: an ultrastructural
analysis of trigeminal root specimens obtained during microvascular decompression surgery.
J
Neurosurg
2002, 96(3):532-543.
Page 14/15
7. Love S, Coakham HB: Trigeminal neuralgia: pathology and pathogenesis.
Brain
2001, 124(Pt
12):2347-2360.
8. Hodaie M, Coello AF: Advances in the management of trigeminal neuralgia.
J Neurosurg Sci
2013,
57(1):13-21.
9. Tohyama S, Hung PS, Zhong J, Hodaie M: Early postsurgical diffusivity metrics for prognostication
of long-term pain relief after Gamma Knife radiosurgery for trigeminal neuralgia.
J Neurosurg
2018,
131(2):539-548.
10. Basser PJ, Mattiello J, LeBihan D: MR diffusion tensor spectroscopy and imaging.
Biophys J
1994,
66(1):259-267.
11. Holdsworth SJ, Skare S, Newbould RD, Guzmann R, Blevins NH, Bammer R: Readout-segmented EPI
for rapid high resolution diffusion imaging at 3T.
Eur J Radiol
2008, 65(1):36-46.
12. Porter DA, Heidemann RM: High resolution diffusion-weighted imaging using readout-segmented
echo-planar imaging, parallel imaging and a two-dimensional navigator-based reacquisition.
Magn
Reson Med
2009, 62(2):468-475.
13. Davis KD, Moayedi M: Central Mechanisms of Pain Revealed Through Functional and Structural MRI.
J Neuroimmune Pharm
2013, 8(3):518-534.
14. Rocca MA, Ceccarelli A, Falini A, Colombo B, Tortorella P, Bernasconi L, Comi G, Scotti G, Filippi M:
Brain gray matter changes in migraine patients with T2-visible lesions - A 3-T MRI study.
Stroke
2006,
37(7):1765-1770.
15. DaSilva AF, Becerra L, Pendse G, Chizh B, Tully S, Borsook D: Colocalized Structural and Functional
Changes in the Cortex of Patients with Trigeminal Neuropathic Pain.
Plos One
2008, 3(10).
16. Gustin SM, Peck CC, Wilcox SL, Nash PG, Murray GM, Henderson LA: Different Pain, Different Brain:
Thalamic Anatomy in Neuropathic and Non-Neuropathic Chronic Pain Syndromes.
J Neurosci
2011,
31(16):5956-5964.
17. Moayedi M, Weissman-Fogel I, Crawley AP, Goldberg MB, Freeman BV, Tenenbaum HC, Davis KD:
Contribution of chronic pain and neuroticism to abnormal forebrain gray matter in patients with
temporomandibular disorder.
Neuroimage
2011, 55(1):277-286.
18. Wiech K, Tracey I: The inuence of negative emotions on pain: Behavioral effects and neural
mechanisms.
Neuroimage
2009, 47(3):987-994.
19. Li M, Yan J, Li S, Wang T, Zhan W, Wen H, Ma X, Zhang Y, Tian J, Jiang G: Reduced volume of gray
matter in patients with trigeminal neuralgia.
Brain Imaging Behav
2017, 11(2):486-492.
20. Blumenfeld-Katzir T, Pasternak O, Dagan M, Assaf Y: Diffusion MRI of Structural Brain Plasticity
Induced by a Learning and Memory Task.
Plos One
2011, 6(6).
21. Zatorre RJ, Fields RD, Johansen-Berg H: Plasticity in gray and white: neuroimaging changes in brain
structure during learning.
Nat Neurosci
2012, 15(4):528-536.
22. Diaz-de-Grenu LZ, Acosta-Cabronero J, Pereira JMS, Pengas G, Williams GB, Nestor PJ: MRI detection
of tissue pathology beyond atrophy in Alzheimer's disease: Introducing T2-VBM.
Neuroimage
2011,
Page 15/15
56(4):1946-1953.
23. Price DD: Neuroscience - Psychological and neural mechanisms of the affective dimension of pain.
Science
2000, 288(5472):1769-1772.
24. DeSouza DD, Hodaie M, Davis KD: Abnormal trigeminal nerve microstructure and brain white matter
in idiopathic trigeminal neuralgia.
Pain
2014, 155(1):37-44.
25. Marinkovic S, Cetkovic M, Gibo H, Todorovic V, Jancic J, Milisavljevic M: Immunohistochemistry of
Displaced Sensory Neurons in the Trigeminal Nerve Root.
Cells Tissues Organs
2010, 191(4):326-
335.
26. Beaulieu C: The basis of anisotropic water diffusion in the nervous system - a technical review.
Nmr
Biomed
2002, 15(7-8):435-455.
27. Silva G, Vieira D, Costa D, Fonseca J: Wallerian degeneration of the pontocerebellar bres secondary
to pontine infarction.
Acta Neurol Belg
2016, 116(4):615-617.
28. De Simone T, Regna-Gladin C, Carriero MR, Farina L, Savoiardo M: Wallerian degeneration of the
pontocerebellar bers.
Am J Neuroradiol
2005, 26(5):1062-1065.
29. DeSouza DD, Moayedi M, Chen DQ, Davis KD, Hodaie M: Sensorimotor and Pain Modulation Brain
Abnormalities in Trigeminal Neuralgia: A Paroxysmal, Sensory-Triggered Neuropathic Pain.
Plos One
2013, 8(6).
30. Wu M, Jiang XF, Qiu J, Fu XM, Niu CS: Gray and white matter abnormalities in primary trigeminal
neuralgia with and without neurovascular compression.
J Headache Pain
2020, 21(1).
31. Kerr FW, Miller RH: The pathology of trigeminal neuralgia. Electron microscopic studies.
Arch Neurol
1966, 15(3):308-319.
32. Hilton DA, Love S, Gradidge T, Coakham HB: Pathological ndings associated with trigeminal
neuralgia caused by vascular compression.
Neurosurgery
1994, 35(2):299-303; discussion 303.
33. Rappaport ZH, Govrin-Lippmann R, Devor M: An electron-microscopic analysis of biopsy samples of
the trigeminal root taken during microvascular decompressive surgery.
Stereotact Funct Neurosurg
1997, 68(1-4 Pt 1):182-186.
34. Love S, Hilton DA, Coakham HB: Central demyelination of the Vth nerve root in trigeminal neuralgia
associated with vascular compression.
Brain Pathol
1998, 8(1):1-11; discussion 11-12.
35. Marinkovic S, Gibo H, Todorovic V, Antic B, Kovacevic D, Milisavljevic M, Cetkovic M: Ultrastructure
and immunohistochemistry of the trigeminal peripheral myelinated axons in patients with neuralgia.
Clin Neurol Neurosurg
2009, 111(10):795-800.
36. Taylor KS, Anastakis DJ, Davis KD: Cutting your nerve changes your brain.
Brain
2009, 132(Pt
11):3122-3133.
37. Davis KD, Taylor KS, Anastakis DJ: Nerve injury triggers changes in the brain.
Neuroscientist
2011,
17(4):407-422.
38. DeSouza DD, Hodaie M, Davis KD: Structural Magnetic Resonance Imaging Can Identify Trigeminal
System Abnormalities in Classical Trigeminal Neuralgia.
Front Neuroanat
2016, 10.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Purpose Previous researches have reported gray and white matter microalterations in primary trigeminal neuralgia (TN) with neurovascular compression (NVC). The central mechanism underlying TN without NVC are unknown but may include changes in specific brain regions or circuitries. This study aimed to investigate abnormalities in the gray matter (GM) and white matter (WM) of the whole brain and the possible pathogenetic mechanism underlying this disease. Methods We analyzed brain morphologic images of TN patients, 23 with NVC (TN wNVC) and 22 without NVC (TN wNVC) compared with 45 healthy controls (HC). All subjects underwent 3T-magnetic resonance imaging and pain scale evaluation. Voxel-based morphometry (VBM) and surface-based morphometry (SBM) were used to investigate whole brain grey matter quantitatively; graph theory was applied to obtain network measures based on extracted DTI data based on DTI data of the whole brains. Sensory and affective pain rating indices were assessed using the visual analog scale (VAS) and short-form McGill Pain Questionnaire (SF-MPQ). Results The VBM and SBM analyses revealed widespread decreases in GM volume and cortical thickness in TN wNVC compared to TN woNVC, and diffusion metrics measures and topology organization changes revealed DTI showed extensive WM integrity alterations. However, above structural changes differed between TN wNVC and TN woNVC, and were related to specific chronic pain modulation mechanism. Conclusion Abnormalities in characteristic regions of GM and WM structural network were found in TN woNVC compared with TN wNVC group, suggesting differences in pathophysiology of two types of TN.
Article
Full-text available
Classical trigeminal neuralgia (TN) is a chronic pain disorder that has been described as one of the most severe pains one can suffer. The most prevalent theory of TN etiology is that the trigeminal nerve is compressed at the root entry zone (REZ) by blood vessels. However, there is significant evidence showing a lack of neurovascular compression (NVC) for many cases of classical TN. Furthermore, a considerable number of patients who are asymptomatic have MR evidence of NVC. Since there is no validated animal model that reproduces the clinical features of TN, our understanding of TN pathology mainly comes from biopsy studies that have limitations. Sophisticated structural MRI techniques including diffusion tensor imaging provide new opportunities to assess the trigeminal nerves and CNS to provide insight into TN etiology and pathogenesis. Specifically, studies have used high-resolution structural MRI methods to visualize patterns of trigeminal nerve-vessel relationships and to detect subtle pathological features at the trigeminal REZ. Structural MRI has also identified CNS abnormalities in cortical and subcortical gray matter and white matter and demonstrated that effective neurosurgical treatment for TN is associated with a reversal of specific nerve and brain abnormalities. In conclusion, this review highlights the advanced structural neuroimaging methods that are valuable tools to assess the trigeminal system in TN and may inform our current understanding of TN pathology. These methods may in the future have clinical utility for the development of neuroimaging-based biomarkers of TN.
Article
Full-text available
Accumulating evidence from brain structural imaging studies has supported that chronic pain could induce changes in brain gray matter volume. However, few studies have focused on the gray matter alterations of Trigeminal neuralgia (TN). In this study, twenty-eight TN patients (thirteen females; mean age, 45.86 years ±11.17) and 28 healthy controls (HC; thirteen females; mean age, 44.89 years ±7.67) were included. Using voxel-based morphometry (VBM), we detected abnormalities in gray matter volume in the TN patients. Based on a voxel-wise analysis, the TN group showed significantly decreased gray matter volume in the bilateral superior/middle temporal gyrus (STG/MTG), bilateral parahippocampus, left anterior cingulate cortex (ACC), caudate nucleus, right fusiform gyrus, and right cerebellum compared with the HC. In addition, we found that the gray matter volume in the bilateral STG/MTG was negatively correlated with the duration of TN. These results provide compelling evidence for gray matter abnormalities in TN and suggest that the duration of TN may be a critical factor associated with brain alterations.
Article
Full-text available
Neurovascular contact is considered a frequent cause of classical trigeminal neuralgia and microvascular decompression with transposition of a blood vessel is preferred over other surgical options in medically refractory patients with classical trigeminal neuralgia. However, the prevalence of neurovascular contact has not been investigated in a representative cohort of patients with classical trigeminal neuralgia based in a neurological setting and using high-quality neuroimaging and blinded evaluation. We aimed to investigate whether presence and degree of neurovascular contact are correlated to pain side in classical trigeminal neuralgia. Consecutive classical trigeminal neuralgia patients with unilateral symptoms were referred to 3.0 T magnetic resonance imaging and included in a cross-sectional study. Magnetic resonance imaging scans were evaluated blindly and graded according to presence and degree of neurovascular contact. Severe neurovascular contact was defined as displacement or atrophy of the trigeminal nerve. A total of 135 patients with classical trigeminal neuralgia were included. Average age of disease onset was 53.0 years (95% confidence interval mean 40.5-55.5) and current age was 60.1 years (95% % confidence interval mean 57.5-62.7). Eighty-two (61%, 95% confidence interval 52-69%) patients were female. Neurovascular contact was prevalent both on the symptomatic and asymptomatic side [89% versus 78%, P = 0.014, odds ratio = 2.4 (1.2-4.8), P = 0.017], while severe neurovascular contact was highly prevalent on the symptomatic compared to the asymptomatic side [53% versus 13%, P < 0.001, odds ratio = 11.6 (4.7-28.9), P < 0.001]. Severe neurovascular contact was caused by arteries in 98%. We conclude that neurovascular contact causing displacement or atrophy of the trigeminal nerve is highly associated with the symptomatic side in classical trigeminal neuralgia as opposed to neurovascular contact in general. Our findings demonstrate that severe neurovascular contact is involved in the aetiology of classical trigeminal neuralgia and that it is caused by arteries located in the root entry zone. © The Author (2014). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oup.com.
Article
Full-text available
Idiopathic trigeminal neuralgia (TN) is classically associated with neurovascular compression (NVC) of the trigeminal nerve at the root entry zone (REZ), but NVC-induced structural alterations are not always apparent on conventional imaging. Previous studies report lower fractional anisotropy (FA) in the affected trigeminal nerves of TN patients using diffusion tensor imaging (DTI). However, it is not known if TN patients have trigeminal nerve abnormalities of mean, radial, or axial diffusivity (MD, RD, AD) - metrics linked to neuroinflammation and edema- or brain white matter (WM) abnormalities. DTI scans were retrospectively analyzed in 18 right-sided TN patients and 18 healthy controls to extract FA, RD, AD and MD from the trigeminal nerve REZ, and used Tract-Based Spatial Statistics (TBSS) to assess brain WM. In patients, the affected trigeminal nerve had lower FA, and higher RD, AD, and MD was found bilaterally compared to controls. Group TBSS results (p<0.05, corrected) showed patients had lower FA and increased RD, MD, and AD in brain WM connecting areas involved in the sensory and cognitive-affective dimensions of pain, attention, and motor functions including the corpus callosum, cingulum, posterior corona radiata, and superior longitudinal fasciculus. These data indicate that TN patients have abnormal tissue microstructure in their affected trigeminal nerves, and as a possible consequence, WM microstructural alterations in the brain. These findings suggest that trigeminal nerve structural abnormalities occur in TN, even if not apparent on gross imaging. Furthermore, MD and RD findings suggest that neuroinflammation and edema may contribute to TN pathophysiology.
Article
Full-text available
Idiopathic trigeminal neuralgia (TN) is characterized by paroxysms of severe facial pain but without the major sensory loss that commonly accompanies neuropathic pain. Since neurovascular compression of the trigeminal nerve root entry zone does not fully explain the pathogenesis of TN, we determined whether there were brain gray matter abnormalities in a cohort of idiopathic TN patients. We used structural MRI to test the hypothesis that TN is associated with altered gray matter (GM) in brain areas involved in the sensory and affective aspects of pain, pain modulation, and motor function. We further determined the contribution of long-term TN on GM plasticity. Cortical thickness and subcortical GM volume were measured from high-resolution 3T T1-weighted MRI scans in 24 patients with right-sided TN and 24 healthy control participants. TN patients had increased GM volume in the sensory thalamus, amygdala, periaqueductal gray, and basal ganglia (putamen, caudate, nucleus accumbens) compared to healthy controls. The patients also had greater cortical thickness in the contralateral primary somatosensory cortex and frontal pole compared to controls. In contrast, patients had thinner cortex in the pregenual anterior cingulate cortex, the insula and the orbitofrontal cortex. No relationship was observed between GM abnormalities and TN pain duration. TN is associated with GM abnormalities in areas involved in pain perception, pain modulation and motor function. These findings may reflect increased nociceptive input to the brain, an impaired descending modulation system that does not adequately inhibit pain, and increased motor output to control facial movements to limit pain attacks.
Article
OBJECTIVE Gamma Knife radiosurgery (GKRS) is an important treatment modality for trigeminal neuralgia (TN). Current longitudinal assessment after GKRS relies primarily on clinical diagnostic measures, which are highly limited in the prediction of long-term clinical benefit. An objective, noninvasive, predictive tool would be of great utility to advance the clinical management of patients. Using diffusion tensor imaging (DTI), the authors’ aim was to determine whether early (6 months post-GKRS) target diffusivity metrics can be used to prognosticate long-term pain relief in patients with TN. METHODS Thirty-seven patients with TN treated with GKRS underwent 3T MRI scans at 6 months posttreatment. Diffusivity metrics of fractional anisotropy, axial diffusivity, radial diffusivity, and mean diffusivity were extracted bilaterally from the radiosurgical target of the affected trigeminal nerve and its contralateral, unaffected nerve. Early (6 months post-GKRS) diffusivity metrics were compared with long-term clinical outcome. Patients were identified as long-term responders if they achieved at least 75% reduction in preoperative pain for 12 months or longer following GKRS. RESULTS Trigeminal nerve diffusivity at 6 months post-GKRS was predictive of long-term clinical effectiveness, where long-term responders (n = 19) showed significantly lower fractional anisotropy at the radiosurgical target of their affected nerve compared to their contralateral, unaffected nerve and to nonresponders. Radial diffusivity and mean diffusivity, correlates of myelin alterations and inflammation, were also significantly higher in the affected nerve of long-term responders compared to their unaffected nerve. Nonresponders (n = 18) did not exhibit any characteristic diffusivity changes after GKRS. CONCLUSIONS The authors demonstrate that early postsurgical target diffusivity metrics have a translational, clinical value and permit prediction of long-term pain relief in patients with TN treated with GKRS. Importantly, an association was found between the footprint of radiation and clinical effectiveness, where a sufficient level of microstructural change at the radiosurgical target is necessary for long-lasting pain relief. DTI can provide prognostic information that supplements clinical measures, and thus may better guide the postoperative assessment and clinical decision-making for patients with TN.
Article
Though classical trigeminal neuralgia (CTN) is frequently caused by neurovascular contact (NVC) at the trigeminal root entry zone (REZ), both anatomical and MRI studies have shown that NVC of the trigeminal nerve frequently occurs in people without CTN. To assess the accuracy of MRI in distinguishing symptomatic from asymptomatic trigeminal NVC, we submitted to high definition MRI the series of CTN patients referred to our outpatient service between June 2011 and January 2013 (n=24), and a similar number of age-matched healthy controls. Two neuroradiologists, blinded to the clinical data, evaluated whether the trigeminal nerve displayed NVC in the REZ or non-REZ, whether it was dislocated by the vessel or displayed atrophy at the contact site, and whether the offending vessel was an artery or a vein. Our data were meta-analyzed with those of all similar studies published from January 1970 to June 2013. In our sample, REZ contact, nerve dislocation and nerve atrophy were independently associated with CTN (p=0.027; p=0.005; p=0.035 respectively). Compared to a rather low sensitivity of each of these items (alone or in combination), their specificity was high. When REZ contact and nerve atrophy coexisted, both specificity and positive predictive value rose to 100%. Meta-analysis showed that REZ NVC was detected in 76% of symptomatic and 17% of asymptomatic nerves (p<0.0001), while anatomical changes were detected in 52% of symptomatic and 9% of asymptomatic nerves (p<0.0001). Trigeminal REZ NVC, as detected by MRI, is highly likely to be symptomatic when it is associated with anatomical nerve changes.
Article
Trigeminal neuralgia (TN), one of the most intense forms of facial pain, has been the subject of great clinical interest. Significant advances have been made in its management, including surgical treatment and imaging of the trigeminal nerve and associated neurovascular compression. The different options for surgical treatment, recent advances in each as well as novel methods of imaging of the trigeminal nerve focusing on diffusion tensor imaging/tractography will be discussed.