Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

INVITED REVIEW

J Neurosurg 139:1471–1479, 2023

The pathophysiology of trigeminal neuralgia:


a molecular review
*Bryan Dong,1 Risheng Xu, MD, PhD,1 and Michael Lim, MD2

Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland; and 2Department of
1

Neurosurgery, Stanford University School of Medicine, Palo Alto, California

OBJECTIVE The goal of this study was to provide a comprehensive overview of the current understanding of molecular
and genetic mechanisms underlying the pathophysiology of trigeminal neuralgia (TN).
METHODS The authors searched PubMed systematically for primary research literature investigating specific molecular
mechanisms from samples derived from patients with TN. The genes/molecules of interest from the selected literature
were then cross-referenced with corresponding studies in animal models of TN.
RESULTS From approximately 345 articles, a total of 12 articles were selected and included in the review, focusing on
ionotropic channel expressivity and mutations, reactive oxygen species expressivity, inflammatory marker expressivity,
and microRNA expressivity. Of the 12 included articles, only 4 had studies completed in other animal models regarding
the corresponding TN mechanism found in humans.
CONCLUSIONS The current literature does not suggest a conclusive disease mechanism for TN in humans. In addition
to neurovascular conflict/compression of the trigeminal nerve, recent studies have indicated that TN may be linked to
inflammatory and reactive oxygen species signaling as well. Recent genetic studies in patients with TN have yet to be
investigated further in animal models.
https://thejns.org/doi/abs/10.3171/2023.2.JNS23274
KEYWORDS trigeminal neuralgia; pain; trigeminal ganglion; reactive oxygen species; TRPA1; NRF2;
neuroinflammation

T
rigeminal neuralgia (TN) is an orofacial pain syn- branches of the trigeminal nerve include the V1 (ophthal-
drome characterized by chronic neuropathic pain mic), V2 (maxillary), and V3 (mandibular) distributions, of
in the trigeminal nerve distribution. TN remains which their afferents supply the dermatomes of the upper,
a significant medical challenge, given that the associated middle, and lower face and jaw, respectively.1 The afferent
pain may profoundly interfere with activities of daily liv- fibers of V1, V2, and V3 converge onto the somas of the
ing.1–3 To date, the pathophysiological mechanisms under- trigeminal ganglion (TG) neurons, which send their cen-
lying TN are yet to be fully understood. TN traditionally tral projections to the trigeminal sensory nuclear complex
has been attributed to physical compression of the trigemi- in the pons.1 TN is the most prevalent neuropathic pain
nal nerve from vascular compression or other underlying disorder affecting the orofacial distribution, and mani-
pathologies.1–3 However, recent evidence points to a role of fests as a severe, piercing pain in the dermatome of the
intrinsic cellular pathways in potentially mediating TN.4,5 trigeminal nerve, most commonly affecting the V2 and
In this review, we will summarize the current literature on V3 distributions.1 TN is classified symptomatically, with
the molecular mechanisms of TN. type I TN defined by intermittent/paroxysmal bursts of
The trigeminal nerve provides motor input to the man- pain, and type II TN defined by constant pain.2 Addition-
dible and somatosensory innervation of the face. The ally, TN is classified based on disease origin, with clas-

ABBREVIATIONS CGRP = calcitonin gene–related peptide; 4-HNE = 4-hydroxynonenal; GABA = γ-aminobutyric acid; GABAAR = GABA type A receptor; IL = interleukin;
ION = infraorbital nerve; miRNA = microRNA; MS = multiple sclerosis; NRF2 = nuclear factor erythroid 2–related factor 2; NVC = neurovascular contact; ROS = reactive
oxygen species; SP = substance P; TG = trigeminal ganglion; TN = trigeminal neuralgia; TNFα = tumor necrosis factor–α; TRP = transient receptor potential; TRPA1 = TRP
ankyrin 1; VIP = vasoactive intestinal peptide.
SUBMITTED February 6, 2023. ACCEPTED February 10, 2023.
INCLUDE WHEN CITING Published online March 31, 2023; DOI: 10.3171/2023.2.JNS23274.
* B.D. and R.X. contributed equally to this work.

©AANS 2023, except where prohibited by US copyright law J Neurosurg Volume 139 • November 2023 1471

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

FIG. 1. Flowchart of search strategy according to the PRISMA 2020 statement. Data added to the PRISMA template (from Page
MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline
for reporting systematic reviews. BMJ 2021;372:n71) under the terms of the Creative Commons Attribution (CC BY 4.0) License
(https://creativecommons.org/licenses/by/4.0/). Figure is available in color online only.

sic TN occurring from neurovascular contact (NVC) at sivity, or other molecular mechanisms were investigated.
the trigeminal nerve root entry zone and secondary TN Articles were searched using the human species filter on
arising from other underlying neurological disorders.3,6,7 PubMed and the following combination of keywords:
However, 7%–29% of patients with TN—those classified trigeminal neuralgia, genetic, molecular, gene, mutation,
with idiopathic TN—do not present with any identifiable channel, and inflammation. The titles and abstracts of ar-
physical abnormalities of the trigeminal nerve.3,8–10 Mo- ticles of interest were individually checked to determine
lecular factors that contribute to orofacial pain in these whether the article was an original clinical study inves-
patients remain poorly characterized. Moreover, NVC at tigating a specific pathophysiological mechanism in pa-
the trigeminal entry zone may also be observed in patients tients with TN. Articles were also manually checked to
who are asymptomatic for TN.11 These findings strongly ensure a discovery cohort of at least 10 patients. Articles
suggest that physical compression of the trigeminal nerve that met these two standards were individually reviewed
alone cannot entirely account for TN. Here we review our on a whole-text basis. Articles regarding trigeminal neu-
current understanding of potential molecular mechanisms ropathic pain were not included.
underlying TN. From the selected articles on patients with TN, the
genes/molecules of interest were identified manually for
each article. We then searched PubMed systematically for
Methods primary research literature on animal models of TN in
The search strategy was conducted according to the which those genes/molecules of interest were investigated.
PRISMA 2020 statement (Fig. 1). We searched PubMed Articles were searched on PubMed using the following
systematically for primary research literature published combination of keywords: trigeminal neuralgia, the gene/
before January 2023 on patients with TN who were part molecule of interest, animal, mouse, and rat. The titles
of a discovery cohort of at least 10 participants in whom and abstracts of potentially related articles were manually
gene/RNA/protein mutations, polymorphisms, expres- checked to verify that the correct genes/molecules were

1472 J Neurosurg Volume 139 • November 2023

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

studied in the same mechanistic context as in the selected application of Cav3.2 channel blockers lessened thermal
article on human TN. nociceptive behaviors.19
From the included human clinical articles, the fol- A wide variety of TRPM family cation channels were
lowing data were extracted: lead author, published year, also shown to have pathological variants related to TN.
discovery cohort size, TN modality in discovery cohort, For instance, a TRPM8-associated mutation discovered in
genes/molecules of significance in the study, study find- 2 siblings with TN lowered the activation threshold, in-
ing, and related animal study on the genes/molecules of creased channel current, and heightened TRPM8 receptor
significance. sensitivity to methanol,20 possibly leading to deleterious
neuronal hyperexcitability and pain. Separately, a TRPM7
mutation was discovered in a family with multigeneration-
Results al TN that increased inward sodium leak current, reduced
From approximately 345 articles yielded in the initial mutant TG neuron activation threshold, and also reduced
search, 17 were identified for manual review. A total of 12 depolarizing resting membrane potential, ultimately lead-
articles were selected and included in the review, focusing ing to aberrant neuronal activity.21 TRPM expressivity
on ionotropic channel expressivity and mutations, reactive may also regulate TN, given that a rat ION constriction
oxygen species (ROS) expressivity, inflammatory marker model showed increased TRPM3 and TRPV4 levels in tri-
expressivity, and microRNA (miRNA) expressivity. Of the geminal spinal subnucleus caudalis.22
12 included articles, only 4 had studies completed in other Currently, a limited number of discovered channel
animal models regarding the corresponding TN mecha- mutations in human TN genetic studies have been vali-
nism found in humans. dated to have abnormal nociceptive processing in animal
models of TN. Notably, GABAAR and Cav3.2 have been
Discussion identified as potential pathological channels for TN im-
plicated in humans as well as validated in mouse mod-
TN as an Ionotropic Channelopathy els.18,19 However, other human polymorphisms detected
Molecular investigations have identified potential ab- in Nav, Cav, voltage-gated potassium channels (Kv), and
normalities in the expressivity or function of cation chan- TRP channels have yet to be established as causal in TN
nels as being associated with TN and other pain syn- transduction (Table 1).17 Furthermore, mutations in iono-
dromes.12 Historically, a nonsense mutation of SCN9A, the tropic channels are exceedingly rare in the TN popula-
gene encoding Nav 1.7, attracted initial interest because it tion. Based on a case report that identified a deleterious
was attributed to a rare congenital loss of nociception in M136V SCN8A mutation in a single patient with TN,23
3 consanguineous families, strongly implicating the pain- Sekula et al. screened 123 patients and were unable to
sensing role of Nav 1.7.13 However, Nav 1.7 and similar identify the M136V in any other patient in their series.24
channel Nav 1.6 variants ultimately have not been estab- Thus, although these recent investigations implicate the
lished as a significant feature in TN pathology, given that pathological role of ionotropic channels in TN, the fact
SCN9A and SCN8A polymorphisms were not correlated remains that germline mutations probably represent a
with patients who have TN.14,15 Moreover, vixotrigine, a small proportion of patients with TN. Increased genetic
Nav 1.7 antagonist used to treat TN, failed to reach its sequencing and routine screening of these patients for
phase II trial primary endpoint.16 The lack of evidence at- genetic mutations will clarify the relative contribution of
tributing Nav 1.7 dysfunction to TN specifically and the channelopathies to this disorder.
failure of vixotrigine imply that orofacial pain in TN is
likely to be mechanistically distinct from that of other pe- TN as an Oxidative Stress Disorder
ripheral neuropathic pain disorders. TN has also been evaluated as a dysfunction of cell
Within the last 5 years there has been increased genetic signaling and transduction networks. Identifying and
evidence suggesting that rare but damaging mutations regulating the centers of these key networks may advance
may predispose a subset of patients with TN to orofacial more effective and robust therapies for TN. Oxidative
pain (Table 1). A genetic study in patients with familial stress refers to an abnormal balance of ROS—derivatives
TN revealed mutations in ionotropic channels, including of oxygen that have oxidizing potential, such as superox-
genes for transient receptor potential (TRP) channels, ide, hydroxyl radicals, and hydrogen peroxide.25 Normally,
voltage-gated potassium channels, and voltage-gated cal- ROS are a product of cellular metabolism and regulate a
cium channels (Fig. 2).17 For instance, exome sequencing range of functions, including cell signaling, survival, and
in patients with TN revealed prominent structural muta- transcription. However, an excess of ROS can lead to the
tions in the γ-aminobutyric acid (GABA)–gated chloride oxidation of proteins, nucleic acids, and lipids, disrupt-
channel GABA type A receptor (GABAAR) and the T- ing normal cellular processes. Oxidative stress has been
type low voltage–gated calcium channel Cav3.2.18 Mutant linked to a spectrum of diseases, such as cancer, diabetes
mice expressing the GABAAR variant displayed mechani- mellitus, and neurodegenerative diseases.26
cal allodynia and pain behavior in the trigeminal distribu- Elevated ROS levels have been observed in chronic
tion, highlighting the possibility of pathological GABA- neuropathic pain syndromes such as diabetic neuropathy,
mediated signaling in trigeminal neuralgia. A recent study chemotherapy-associated neuropathy, spinal cord injury,
also independently confirmed the role of mutant Cav3.2 in and, recently, TN.27 Recently, Vasavda et al. discovered
a mouse model of TN; Cav3.2 expression was elevated in that CSF from patients with TN had increased 4-hy-
a mouse model of infraorbital nerve (ION) ligation, and droxynonenal (4-HNE) and malondialdehyde, which are

J Neurosurg Volume 139 • November 2023 1473

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

TABLE 1. List of human genetic/molecular studies in patients with TN and related studies in animal models
Original
Study, Authors Discovery Genes/Molecules of
& Year Cohort Significance Study Findings Related Animal Study
Vasavda et al., 13 pts w/ TRPA1, NRF2 CSF: pts w/ TN had upregulated ROS ROS signaling through TRPA1 contrib-
202228 primary TN levels mediating noxious, inflammatory utes to trigeminal neuropathic pain,
cascade along TRPA1 axis NRF2 activation alleviates TRPA1-
induced nociception24
Hale et al., 1188 pts w/ TN Genome-wide association 5 single-nucleotide polymorphisms at in- None
202264 study tergenic locus on chromosome 1p22.2,
variant on chromosome 5q35.1
Romero et al., 48 pts w/ SCN8A, BDNF, COMT, GCH1 Polymorphisms in SCN8A, BDNF, COMT, None
202115 classic or & GCH1 were not associated w/ TN
idiopathic TN onset or orofacial pain intensity
Sekula et al., 123 pts w/ clas- SCN8A M136V mutation of SCN8A was not identi- None
202124 sic TN fied in any pts in discovery cohort
Di Stefano et 11 pts w/ SCN2A, SCN3A, SCN5A, Sequencing revealed rare, potentially None
al., 202017 classic or SCN7A, SCN9A, SCN10A, pathogenic mutations of 36 genes
idiopathic TRPA1, TRPC6, TRPM2, encoding voltage-dependent Na+, K+,
familial TN TRPM3, TRPM4, TRPM7, Ca2+, C1–, nonspecific ligand–gated
TRPM8, TRPS1, TRPV4, channels, & gap junction channels
TRPV5, TRPV6, KCNA5,
KCNC3, KCND2, KCNH2,
KCNH7, KCNJ6, KCNK17,
KCNS2, KCNV1, CACNA1A,
CACNA1D, CACNA1G,
CACNA1I, CACNA1S,
CACNB1, CLCN1, CLCN2,
CLIC5, GJB5
Dong et al., 290 pts w/ mul- GABRG1, CACNA1H, TRAK1 Genomic screening of pts w/ TN revealed Mutant GABA AR contributes to me-
202018 timodal TN rare, deleterious GABRG1, CACNA1H, chanical allodynia sensing in mice,14
& TRAK1 variants mutant Cav3.2 contributes to thermal
allodynia sensing in mice15
Li et al., 28 pts w/ clas- miR-132-3p, miR-146b-5p,miR-132-3p, miR-146b-5p, miR-155-5p, None
202033 sic TN miR-155-5p, & miR-384 & miR-384 were highly upregulated in
serum of pts w/ TN
Costa et al., 48 pts w/ clas- SCN9A, NTRK1 Polymorphisms in SCN9A & NTRK1 were None
201914 sic TN not associated w/ TN onset or orofacial
pain intensity
Qin et al., 20 pts w/ CGRP, SP, VIP, & β-endorphin Increased levels of CGRP, SP, & VIP & CGRP mRNA was upregulated in rat TG
201647 primary TN lowered levels of β-endorphin in CSF after ION constriction,60 CGRP & SP
of pts w/ TN levels were increased in rat TG after
ION constriction,61 greater no. of SP
immunopositive cells in rat TG after
ION constriction,62 heat hyperalgesia
was reduced after administration of
SP receptor NK1 antagonist63
Cui et al., 280 pts w/ idio- SLC6A4 SLC6A4 short-short alleles were more None
201466 pathic TN expressed in pts w/ TN, & associated
w/ higher levels of pain
Siqueira et al., 10 pts w/ TN SCN3A, SCN9A, SCN10A Increased expressivity of Nav1.3 & Nav1.7 None
2009 65 mRNA in gingival tissue of pts w/ TN
Strittmatter et 16 pts w/ idio- Epinephrine, norepinephrine, Increased levels of SP & lowered levels Same studies regarding SP
al., 199648 pathic TN dopamine, VMA, HVA, SP, of norepinephrine, somatostatin, VMA,
5-HIAA, somatostatin HVA, & 5-HIAA in CSF of pts w/ TN
5-HIAA = 5-hydroxyindoleacetic acid; HVA = homovanillic acid; mRNA = messenger RNA; pts = patients; VMA = vanillylmandelic acid.

1474 J Neurosurg Volume 139 • November 2023

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

FIG. 2. Overview of genetic and molecular pathways implicated in orofacial pain signaling in TG neurons in human and animal
models of TN. NVC or secondary lesions impinge on the trigeminal nerve root, causing mechanical injury to the nerve. Mechanical
injury can cause ROS production, leading to activation of the TRPA1 channels, which are regulated by the NRF2 axis. Neuroin-
flammation, which may be downstream of mechanical injury or secondary to autoimmune disease such as MS, may also increase
neuropathic pain. Rare but damaging mutations of genes encoding T-type low-voltage–gated calcium channel (Cav3.2) and
GABA AR may lower activation potential and cause hyperexcitability in TG neurons. Figure is available in color online only.

ROS products of lipid peroxidation.28 These ROS interact 5p signaling is directly related with noxious sensing in an
with the TRP ankyrin 1 (TRPA1) channel, a nonselective animal model of TN remains to be tested.
cation channel that is selectively expressed in nociceptive These studies indicate that TRPA1 may be a potential
mammalian small-diameter dorsal root ganglion and TG therapeutic agent for TN by targeting aberrant oxidative
neurons, increasing its response to nerve injury in a mouse stress signaling. However, challenges remain in develop-
model of trigeminal nerve compression.29,30 This work ing TRPA1-directed therapeutics, given that all TRPA1
builds on prior studies by Trevisani et al. and Andersson antagonists thus far have failed in animal or clinical tri-
et al., who found that application of 4-HNE produced de- als.34–36 For instance, the TRPA1 inhibitors GRC17536
polarizing currents in rat and mouse dorsal root ganglion and ODM-108 were terminated at the phase II and phase I
neurons and increased their sensitivity to mechanical allo- stages, respectively, due to poor pharmacokinetics in both
dynia; inhibiting TRPA1 attenuated the 4-HNE–induced instances.35,36 Vasavda et al. circumvented these drawbacks
depolarization and reduced pain behavior.31,32 Vasavda et by targeting NRF2 pathways upstream of TRPA1. NRF2
al. further demonstrated that pharmacological or genetic expression levels are strictly regulated at baseline by Kelch-
activation of nuclear factor erythroid 2–related factor 2 like ECH–associated protein 1–mediated tagging and
(NRF2), a key transcriptional driver of the cellular anti- ubiquitination.37 By pretreating mice with sulforaphane,
oxidant response, was important in decreasing mechanical a Kelch-like ECH–associated protein 1 inhibitor, NRF2
and cold allodynia in the mouse maxillary nerve constric- levels were upregulated and 4-HNE and malondialdehyde
tion model (Fig. 2).28 levels were diminished in TG neurons, and the intensity
Interestingly, an independent transcriptomic study by of mechanical and cold allodynia was lessened after max-
Li et al. investigated the miRNA serum profile of patients illary nerve constriction. Additionally, the authors found
with TN and found that 4 miRNAs were expressed at sig- that two compounds, exemestane and JQ-1, could induce
nificantly higher levels in patients with TN.33 One of these, a transcriptional landscape similar to NRF2, possibly by
miR-155–5p, was shown to target NRF2 transcripts and exemestane activating the canonical NRF2 network and
downregulate cellular NRF2 levels, indicating a possible JQ-1 driving antioxidant signaling parallel to NRF2.37 Ap-
endogenous pathway by which NRF2-driven oxidative re- plication of exemestane or JQ-1 again decreased the mag-
sponse is suppressed in TN. However, whether miR-155– nitude of mechanical and cold allodynia in a mouse model

J Neurosurg Volume 139 • November 2023 1475

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

of TN. Importantly, exemestane and JQ-1 are appealing for matory regulators may provide an alternative strategy in
TN therapy because exemestane is already approved as a treating TN. Still, despite many clinical and basic research
breast cancer treatment, and analogs to JQ-1 are currently studies linking inflammatory signaling and TN, more re-
being investigated in clinical trials for cancer therapy.37 search is needed to delineate the causes and interactions of
By considering TN as a manifestation of oxidative inflammation in different TN subtypes.
stress, at least two potential therapeutic avenues emerge:
inhibiting the direct transduction of ROS nociceptive sig- Shortcomings of Animal Models of TN
nals at the TRPA1 interface, or promoting the antioxidant Although rare historical experiments have assessed
response from the global regulator NRF2. Future stud- physiological features of trigeminal nerve tissue from pa-
ies into the genomic and signaling dynamics of oxidative tients with TN and found gliosis and demyelination,57,58
stress feedback in pain models may yield more effective there have been no modern molecular studies character-
and specific therapies for treating TN. izing the dynamics of inflammation in neuronal tissue
from patients with TN, due to the ethical limitations of
TN as a Neuroinflammatory Disorder procuring trigeminal nerve samples from live patients.
Orofacial pain in TN may also be affected by inflam- This paucity of trigeminal nerve tissue from patients with
matory signaling pathways. Multiple sclerosis (MS) is a TN represents a major hurdle in identifying the role of
well-described autoimmune disease in which central de- intracellular/molecular mediators of pain signal transduc-
myelination and plaque formation contribute to a range of tion. Thus, to further interrogate the role of inflammation
motor and sensory neurological deficits.38–40 A small but and/or other cellular mechanisms in TN, improved trans-
well-delineated subgroup of patients with TN, ranging lational animal models are necessary to establish causal
from 2.4% to 8%, have concomitant MS.41,42 Multiple MRI relationships between human correlative studies and true
studies have indicated that TN secondary to MS is asso- TN pathophysiology.
ciated with demyelinating plaques located in the pontine Current animal models that attempt to characterize TN
region of the primary afferents of the trigeminal nerve be- have numerous drawbacks, complicating efforts to bridge
tween the root entry zone and the trigeminal nucleus.43,44 It TN from bench to bedside. A majority of rodent models
is still unknown how these central MS-associated plaques of TN apply mechanical ligation to the ION or suborbital
contribute to TN—whether demyelination promotes aber- branches of the V2 branch of the trigeminal nerve to stim-
rant noxious activity directly (e.g., aberrant firing of nerves ulate the painful features of TN.59 However, these constric-
compared to ephaptic coupling), or through indirect, in- tion injury models are more similar to painful trigeminal
flammatory pathways. neuropathy, representing a continuous mechanical noxious
The interplay between demyelination, inflammatory stimulus and failing to capture the paroxysms common in
cues, and TN is not yet fully characterized; however, TN. Additionally, ligation studies do not represent the full
neuroinflammation may play a role in orofacial pain.45 spectrum of pathological agents precipitating TN, given
Multiple studies in patients with TN have identified up- that idiopathic TN occurs independently of any physical
regulated levels of inflammatory biomarkers in CSF and injury. Furthermore, modeling TN with only mechanical
blood serum, including substance P (SP), calcitonin gene– insults fails to capture the deleterious cellular intrinsic and
related peptide (CGRP), and vasoactive intestinal peptide extrinsic signaling evidenced in genetic TN studies.
(VIP),46–48 as well as endogenous regulators of inflamma- Fundamentally, ligation of V2 subbranches may not ac-
tion like tumor necrosis factor–α (TNFα) and inflamma- curately depict the underlying determinant of primary TN,
tory cytokine interleukin-6 (IL-6).49,50 Indeed, enhanced- which occurs due to compression at the trigeminal nerve
activity animal studies show that overexpression of TNFα root entry zone. Indeed, constriction of trigeminal nerve
in rat TG neurons increased sensitivity to mechanical no- branches more distal to TG neurons may overlook central
ciception,51 and a rat ION constriction model showed up- transduction and integration aberrations present in TN.
regulated levels of IL-6.52 Manipulation of these key con- Recent studies in rats have attempted to address this short-
trollers of neuroinflammation may be leveraged to treat coming by compressing the trigeminal nerve at the root
TN. For instance, downregulation of long noncoding RNA entry zone,60,61 representing a more facile model compared
Gm14461, an endogenous positive regulator of TNFα, to mice, given the larger animal model. However, unlike
IL-6, and another inflammatory cytokine (IL-1β) in TG corresponding human studies,46–48 trigeminal nerve root
neurons decreased mechanical nociception sensitivity in compression in rats resulted in reduced levels of SP and
a mouse model of TN, whereas overexpressing Gm14461 CGRP in the medullary dorsal horn,60 again indicating the
showed the opposite effect.53 possible insufficiency of neurocompressive animal models
Mechanistically, inflammatory mediators have also in accurately capturing TN etiology. Thus, basic research
been shown to regulate expression of channels implicated models of TN are currently insufficient in replicating the
in noxious signaling, with TNFα contributing to an up- full breadth of etiological factors driving this disease. Mo-
regulation in noxious mechanosensitive receptors P2X lecular TN studies that use ION or suborbital nerve liga-
purinoceptor 3 (P2X3) and P2X purinoceptor 4 (P2X4) in tion as their disease model cannot be directly translated to
TG neurons, among others.54,55 Likewise, IL-6 increases clinical application without careful consideration, because
TG expression of Nav 1.3 and mechanosensitive chan- these frameworks may only address neuropathic pain that
nel Piezo2 in the TG neurons in rodent models of TN.52,56 is parallel to but ultimately not TN. Ultimately, more com-
Thus, identifying and mediating critical pathway elements prehensive and global models of TN need to be developed
downstream of TNFα, IL-6, and similar neuroinflam- to provide greater translational advantages.
1476 J Neurosurg Volume 139 • November 2023

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

Future Directions and Unanswered Questions 5. Smith CA, Paskhover B, Mammis A. Molecular mechanisms
The most effective current surgical treatment for TN re- of trigeminal neuralgia:​a systematic review. Clin Neurol
Neurosurg. 2021;​200:​106397.
mains microvascular decompression, which is predicated 6. Jones MR, Urits I, Ehrhardt KP, et al. A comprehensive
on resolving the offending NVC.1,3,7 However, it remains review of trigeminal neuralgia. Curr Pain Headache Rep.
unclear whether the analgesic effect of decompression 2019;​23(10):​74.
results from the removal of physical contact between the 7. So RJ, Kalluri AL, Zhu S, et al. Multiple vessel compression
vessel and nerve or from molecular mechanisms second- of the trigeminal nerve is associated with worse outcomes
ary to NVC. Indeed, approximately one-quarter of patients in trigeminal neuralgia after microvascular decompression.
with TN still experience persistent pain years after micro- Neurosurgery. Published online December 30, 2022. doi:​
vascular decompression,62 suggesting that nonmechanical 10.1227/neu.0000000000002323
8. Meaney JF, Eldridge PR, Dunn LT, Nixon TE, Whitehouse
pathways may still be significant in compressive forms GH, Miles JB. Demonstration of neurovascular compression
of TN. Thus, when considering the possible molecular in trigeminal neuralgia with magnetic resonance imaging.
mechanisms of TN, it is important to consider the types of Comparison with surgical findings in 52 consecutive opera-
TN being investigated. It is unknown whether the differ- tive cases. J Neurosurg. 1995;​83(5):​799-805.
ent molecular mechanisms discussed above each leads to 9. Lee A, McCartney S, Burbidge C, Raslan AM, Burchiel KJ.
a distinct form of TN, or if a combination of these mecha- Trigeminal neuralgia occurs and recurs in the absence of
nisms generates all forms of TN. For instance, Vasavda et neurovascular compression. J Neurosurg. 2014;​120(5):​1048-
1054.
al.’s study only investigated ROS levels in CSF of patients 10. Xu R, Materi J, Raj D, et al. Internal neurolysis versus
with classic TN.28 Whether the noxious ROS/TRPA1 sig- intraoperative glycerin rhizotomy for trigeminal neuralgia. J
naling axis is present in all forms of TN, or only where Neurosurg. 2022;​138(1):​270-275.
there is NVC remains unclear. Additionally, although 11. Antonini G, Di Pasquale A, Cruccu G, et al. Magnetic
it is plausible that a subset of secondary TN arises from resonance imaging contribution for diagnosing symptomatic
inflammation underlying pathologies such as MS,63 no neurovascular contact in classical trigeminal neuralgia:​a
conclusive studies have shown that TN is mechanistically blinded case-control study and meta-analysis. Pain. 2014;​
downstream of MS. Finally, idiopathic TN may arise from 155(8):​1464-1471.
12. Raouf R, Quick K, Wood JN. Pain as a channelopathy. J Clin
exceedingly rare channel mutations in singular cases,23 but Invest. 2010;​120(11):​3745-3752.
the inconsistency of such findings poses a challenge for 13. Cox JJ, Reimann F, Nicholas AK, et al. An SCN9A channelo-
effective therapeutic management.24 Future studies of TN pathy causes congenital inability to experience pain. Nature.
need to address the differing pathological mechanisms re- 2006;​444(7121):​894-898.
garding the mode of TN. 14. Costa GMF, Rocha LPC, Siqueira SRDT, Moreira PR,
Almeida-Leite CM. No association of polymorphisms in
Nav1.7 or nerve growth factor receptor genes with trigeminal
Conclusions neuralgia. Pain Med. 2019;​20(7):​1362-1369.
TN is classically described as a neuropathic pain condi- 15. Romero J, Costa GMF, Rocha LPC, Siqueira S, Moreira
tion arising from the compression of the trigeminal nerve. PR, Almeida-Leite CM. Polymorphisms of Nav1.6 sodium
We have summarized the current literature on genetic channel, brain-derived neurotrophic factor, catechol-O-meth-
yltransferase and guanosine triphosphate cyclohydrolase 1
studies and basic research models of TN in which the like- genes in trigeminal neuralgia. Clin Neurol Neurosurg. 2021;​
ly role of various molecular systems associated with and/ 208:​106880.
or precipitating TN is implicated. Although a wealth of 16. Zakrzewska JM, Palmer J, Morisset V, et al. Safety and
evidence points to germline mutations in causing a sub- efficacy of a Nav1.7 selective sodium channel blocker in
set of TN, challenges remain in effective, channel-based patients with trigeminal neuralgia:​a double-blind, placebo-
therapeutics because the true prevalence of these rare but controlled, randomised withdrawal phase 2a trial. Lancet
damaging mutations remains unclear. Thus, focusing on Neurol. 2017;​16(4):​291-300.
inflammation, oxidative stress, and other downstream 17. Di Stefano G, Yuan JH, Cruccu G, Waxman SG, Dib-Hajj
SD, Truini A. Familial trigeminal neuralgia—a systematic
molecular pathways that give rise to TN may provide an clinical study with a genomic screen of the neuronal elec-
alternative translational approach. Further molecular in- trogenisome. Cephalalgia. 2020;​40(8):​767-777.
vestigations are integral in uncovering the underlying fac- 18. Dong W, Jin SC, Allocco A, et al. Exome sequencing impli-
tors of TN and laying the groundwork for developing novel cates impaired GABA signaling and neuronal ion transport
remedies for this disease. in trigeminal neuralgia. iScience. 2020;​23(10):​101552.
19. Gambeta E, Gandini MA, Souza IA, Zamponi GW. Ca V 3.2
calcium channels contribute to trigeminal neuralgia. Pain.
References 2022;​163(12):​2315-2325.
1. Gambeta E, Chichorro JG, Zamponi GW. Trigeminal neural- 20. Gualdani R, Yuan JH, Effraim PR, et al. Trigeminal neural-
gia:​an overview from pathophysiology to pharmacological gia TRPM8 mutation:​enhanced activation, basal [Ca2+]i and
treatments. Mol Pain. 2020;​16:​1744806920901890. menthol response. Neurol Genet. 2021;​7(1):​e550.
2. Yadav YR, Nishtha Y, Sonjjay P, Vijay P, Shailendra R, Yatin K. 21. Gualdani R, Gailly P, Yuan JH, et al. A TRPM7 mutation
Trigeminal neuralgia. Asian J Neurosurg. 2017;​12(4):​585-597. linked to familial trigeminal neuralgia:​omega current and
3. Cruccu G, Finnerup NB, Jensen TS, et al. Trigeminal neu- hyperexcitability of trigeminal ganglion neurons. Proc Natl
ralgia:​new classification and diagnostic grading for practice Acad Sci U S A. 2022;​119(38):​e2119630119.
and research. Neurology. 2016;​87(2):​220-228. 22. Yang KY, Kim MJ, Ju JS, et al. Antinociceptive effects of
4. Xu R, Xie ME, Jackson CM. Trigeminal neuralgia:​current botulinum toxin type A on trigeminal neuropathic pain. J
approaches and emerging interventions. J Pain Res. 2021;​14:​ Dent Res. 2016;​95(10):​1183-1190.
3437-3463. 23. Tanaka BS, Zhao P, Dib-Hajj FB, et al. A gain-of-function

J Neurosurg Volume 139 • November 2023 1477

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

mutation in Nav1.6 in a case of trigeminal neuralgia. Mol tiple sclerosis:​lesion localization with magnetic resonance
Med. 2016;​22(1):​338-348. imaging. Neurology. 1997;​49(4):​1142-1144.
24. Sekula RF, Deeley K, Denwood H, Vieira AR. Gain-of-func- 44. Cruccu G, Biasiotta A, Di Rezze S, et al. Trigeminal neural-
tion mutation Met136Val in SCN8A may not be a common gia and pain related to multiple sclerosis. Pain. 2009;​143(3):​
cause of trigeminal neuralgia. Mol Genet Genomic Med. 186-191.
2021;​9(2):​e1587. 45. Xu R, Nair SK, Shah PP, et al. A potential role for steroids in
25. Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotro- acute pain management in patients with trigeminal neuralgia.
pic physiological signalling agents. Nat Rev Mol Cell Biol. World Neurosurg. 2022;​167:​e1291-e1298.
2020;​21(7):​363-383. 46. Strittmatter M, Grauer M, Isenberg E, et al. Cerebrospinal
26. Pizzino G, Irrera N, Cucinotta M, et al. Oxidative stress:​ fluid neuropeptides and monoaminergic transmitters in
harms and benefits for human health. Oxid Med Cell Longev. patients with trigeminal neuralgia. Headache. 1997;​37(4):​
2017;​2017:​8416763. 211-216.
27. Carrasco C, Naziroǧlu M, Rodríguez AB, Pariente JA. 47. Qin ZL, Yang LQ, Li N, et al. Clinical study of cerebrospi-
Neuropathic pain:​delving into the oxidative origin and the nal fluid neuropeptides in patients with primary trigeminal
possible implication of transient receptor potential channels. neuralgia. Clin Neurol Neurosurg. 2016;​143:​111-115.
Front Physiol. 2018;​9:​95. 48. Strittmatter M, Grauer M, Isenberg E, et al. Substance P,
28. Vasavda C, Xu R, Liew J, et al. Identification of the NRF2 somatostatin and monoaminergic transmitters in the cere-
transcriptional network as a therapeutic target for trigeminal brospinal fluid of patients with chronic idiopathic trigeminal
neuropathic pain. Sci Adv. 2022;​8(31):​eabo5633. neuralgia. Article in German. Schmerz. 1996;​10(5):​261-268.
29. Kobayashi K, Fukuoka T, Obata K, et al. Distinct expression 49. Ericson H, Abu Hamdeh S, Freyhult E, et al. Cerebrospinal
of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary af- fluid biomarkers of inflammation in trigeminal neuralgia
ferent neurons with adelta/c-fibers and colocalization with trk patients operated with microvascular decompression. Pain.
receptors. J Comp Neurol. 2005;​493(4):​596-606. 2019;​160(11):​2603-2611.
30. Kwan KY, Allchorne AJ, Vollrath MA, et al. TRPA1 con- 50. Liu MX, Zhong J, Xia L, Dou NN, Li ST. A correlative analy-
tributes to cold, mechanical, and chemical nociception but is sis between inflammatory cytokines and trigeminal neuralgia
not essential for hair-cell transduction. Neuron. 2006;​50(2):​ or hemifacial spasm. Neurol Res. 2019;​41(4):​335-340.
277-289. 51. Lu ZY, Fan J, Yu LH, Ma B, Cheng LM. The up-regulation
31. Andersson DA, Gentry C, Moss S, Bevan S. Transient recep- of TNF-α maintains trigeminal neuralgia by modulating
tor potential A1 is a sensory receptor for multiple products of MAPKs phosphorylation and BKCa channels in trigeminal
oxidative stress. J Neurosci. 2008;​28(10):​2485-2494. nucleus caudalis. Front Cell Neurosci. 2021;​15:​764141.
32. Trevisani M, Siemens J, Materazzi S, et al. 4-Hydroxynone- 52. Liu M, Li Y, Zhong J, Xia L, Dou N. The effect of IL-6/
nal, an endogenous aldehyde, causes pain and neurogenic in- Piezo2 on the trigeminal neuropathic pain. Aging (Albany
flammation through activation of the irritant receptor TRPA1. NY). 2021;​13(10):​13615-13625.
Proc Natl Acad Sci U S A. 2007;​104(33):​13519-13524. 53. Xu M, Yan Y, Zhu M, Wang Z, Zhang X, Zhang D. Effects
33. Li X, Wang D, Zhou J, Yan Y, Chen L. Evaluation of circulat- of long non-coding RNA Gm14461 on pain transmission in
ing microRNA expression in patients with trigeminal neu- trigeminal neuralgia. J Inflamm (Lond). 2020;​17(1):​1.
ralgia:​an observational study. Medicine (Baltimore). 2020;​ 54. Koizumi M, Asano S, Furukawa A, et al. P2X3 receptor
99(48):​e22972. upregulation in trigeminal ganglion neurons through TNFα
34. Schwarz MG, Namer B, Reeh PW, Fischer MJM. TRPA1 and production in macrophages contributes to trigeminal neuro-
TRPV1 antagonists do not inhibit human acidosis-induced pathic pain in rats. J Headache Pain. 2021;​22(1):​31.
pain. J Pain. 2017;​18(5):​526-534. 55. Liao JY, Zhou TH, Chen BK, Liu ZX. Schwann cells and
35. Koivisto A, Jalava N, Bratty R, Pertovaara A. TRPA1 antago- trigeminal neuralgia. Mol Pain. 2020;​16:​1744806920963809.
nists for pain relief. Pharmaceuticals (Basel). 2018;​11(4):​117. 56. Liu MX, Zhong J, Xia L, Dou NN, Li ST. IL-6 contributes to
36. Koivisto AP, Belvisi MG, Gaudet R, Szallasi A. Advances in Nav1.3 up-regulation in trigeminal nerve following chronic
TRP channel drug discovery:​from target validation to clin- constriction injury. Neurol Res. 2020;​42(6):​504-514.
ical studies. Nat Rev Drug Discov. 2022;​21(1):​41-59. 57. Love S, Gradidge T, Coakham HB. Trigeminal neuralgia due
37. Itoh K, Wakabayashi N, Katoh Y, et al. Keap1 represses to multiple sclerosis:​ultrastructural findings in trigeminal
nuclear activation of antioxidant responsive elements by Nrf2 rhizotomy specimens. Neuropathol Appl Neurobiol. 2001;​
through binding to the amino-terminal Neh2 domain. Genes 27(3):​238-244.
Dev. 1999;​13(1):​76-86. 58. Hilton DA, Love S, Gradidge T, Coakham HB. Pathologi-
38. Hauser SL, Oksenberg JR. The neurobiology of multiple scle- cal findings associated with trigeminal neuralgia caused by
rosis:​genes, inflammation, and neurodegeneration. Neuron. vascular compression. Neurosurgery. 1994;​35(2):​299-303.
2006;​52(1):​61-76. 59. Fried K, Hansson PT. Animal models of trigeminal neural-
39. Bitsch A, Schuchardt J, Bunkowski S, Kuhlmann T, Brück gia:​a commentary. Mol Pain. 2020;​16:​1744806920980538.
W. Acute axonal injury in multiple sclerosis. Correlation with 60. Luo DS, Zhang T, Zuo CX, et al. An animal model for
demyelination and inflammation. Brain. 2000;​123(Pt 6):​1174- trigeminal neuralgia by compression of the trigeminal nerve
1183. root. Pain Physician. 2012;​15(2):​187-196.
40. Selmaj KW, Raine CS. Tumor necrosis factor mediates my- 61. Luo D, Lin R, Luo L, et al. Glial plasticity in the trigeminal
elin and oligodendrocyte damage in vitro. Ann Neurol. 1988;​ root entry zone of a rat trigeminal neuralgia animal model.
23(4):​339-346. Neurochem Res. 2019;​44(8):​1893-1902.
41. Jensen TS, Rasmussen P, Reske-Nielsen E. Association of 62. Cruccu G. Trigeminal neuralgia. Continuum (Minneap
trigeminal neuralgia with multiple sclerosis:​clinical and Minn). 2017;​23(2, Selected Topics in Outpatient Neurology):​
pathological features. Acta Neurol Scand. 1982;​65(3):​182-189. 396-420.
42. Ferraro D, Annovazzi P, Moccia M, et al. Characteristics and 63. Di Stefano G, Maarbjerg S, Truini A. Trigeminal neuralgia
treatment of multiple sclerosis-related trigeminal neuralgia:​ secondary to multiple sclerosis:​from the clinical picture to
an Italian multi-centre study. Mult Scler Relat Disord. 2020;​ the treatment options. J Headache Pain. 2019;​20(1):​20.
37:​101461. 64. Hale AT, Akinnusotu O, He J, et al. Genome-wide associa-
43. Gass A, Kitchen N, MacManus DG, Moseley IF, Hennerici tion study reveals genetic risk factors for trigeminal neural-
MG, Miller DH. Trigeminal neuralgia in patients with mul- gia. Neurosurgery. 2022;​68(suppl 1):​36-37.

1478 J Neurosurg Volume 139 • November 2023

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC
Dong et al.

65. Siqueira SR, Alves B, Malpartida HM, Teixeira MJ, Siqueira point inhibitors, and Checkpoints for Neuro-Inflammation; and is
JT. Abnormal expression of voltage-gated sodium channels a nonresearch consultant for Stryker’s Data and Safety Monitoring
Nav1.7, Nav1.3 and Nav1.8 in trigeminal neuralgia. Neurosci- Board—Cellularity.
ence. 2009;​164(2):​573-577.
66. Cui W, Yu X, Zhang H. The serotonin transporter gene Author Contributions
polymorphism is associated with the susceptibility and the Conception and design: all authors. Analysis and interpretation of
pain severity in idiopathic trigeminal neuralgia patients. J data: all authors. Drafting the article: all authors. Critically revis-
Headache Pain. 2014;​15(1):​42. ing the article: all authors. Reviewed submitted version of manu-
script: all authors. Approved the final version of the manuscript
on behalf of all authors: Lim. Statistical analysis: all authors.
Disclosures Administrative/technical/material support: all authors.
Dr. Lim reported grants from Biohaven outside the submit-
ted work; in addition, he has a patent for NRF2 Pathway for Correspondence
TN pending. He also received research support from Arbor, Michael Lim: Stanford University School of Medicine, Palo Alto,
BMS, Accuray, Biohaven, and Urogen; he is a consultant for CA. mklim@stanford.edu.
VBI, InCephalo Therapeutics, Merck, Pyramid Bio, Insightec,
Biohaven, Sanianoia, Hemispherian, Novocure, Noxxon, InCando,
Century Therapeutics, CraniUs, MediFlix, Stryker, and XSense;
he is a shareholder in Egret Therapeutics; has a patent for Focused
radiation + checkpoint inhibitors, Local chemotherapy + check-

J Neurosurg Volume 139 • November 2023 1479

Brought to you by PUC - RJ Pontificia Universidade Catolica do Rio de Janeiro | Unauthenticated | Downloaded 04/01/24 04:54 PM UTC

You might also like