Skip to main content

Trigeminal ganglion itself can be a viable target to manage trigeminal neuralgia

Abstract

Excruciating trigeminal neuralgia (TN) management is very difficult and severely affects the patient’s quality of life. Earlier studies have shown that the trigeminal ganglion (TG) comprises several receptors and signal molecules that are involved in the process of peripheral sensitization, which influences the development and persistence of neuropathic pain. Targeting TG can modulate this sensitization pathway and mediate the pain-relieving effect. So far,there are few studies in which modulation approaches to TG itself have been suggested so far. “Trigeminal ganglion modulation” and “trigeminal neuralgia” were used as search phrases in the Scopus Index and PubMed databases to discover articles that were pertinent to the topic. In this review, we address the role of the trigeminal ganglion in TN and underlying molecules and neuropeptides implicated in trigeminal pain pathways in processing pathological orofacial pain. We also reviewed different modulation approaches in TG for TN management. Furthermore, we discuss the prospect of targeting trigeminal ganglion to manage such intractable pain.

Peer Review reports

Background

Trigeminal neuralgia (TN), commonly referred to as tic douloureux, is a severe facial condition marked by intense, quickly passing stabbing pain attacks that are elicited by stimuli [1]. The trigger factors of TN include routine activities like chewing, talking, and brushing. Small sensory-sensitive zones are where these triggers are situated. The sites of a sensory trigger and the sites of pain, however, are not always the same [2]. The major pathophysiological mechanisms of TN include compression of the trigeminal nerve root, primary demyelinating disease, and central pain-related circuit sensitization and dysfunction. The hyperactivity or aberrant discharge from the injured nerve roots, ganglion, and trigeminal nucleus in the brainstem is speculated to be accountable for the pain. Medication, surgery, and complementary treatment approaches are offered. Medical pharmacotherapy is the preferred initial treatment for TN. Carbamazepine and oxcarbazepine (sodium channel blockers) are the first-line medications. Other cornerstones of treatment are drugs like tricyclic antidepressants and anticonvulsants [3]. Despite being the most prevalent neuralgia, it is a rare condition. TN still presents difficulties for healthcare professionals as efficient, safe, and well-established therapies cannot be undertaken till a precise diagnosis is made. Dental pain, migraine, cluster headaches, and temporomandibular joint disorders are all common misdiagnoses [4, 5]. Nerve injections with local anesthetics or steroids may provide temporary pain relief for certain people, but most of the time, the relief is just momentary. In the past, neurosurgical destructive and ablative procedures like neurotomies, neurolysis, and surgical decompression have been frequently utilized. These methods could, however, lead to issues like the emergence of delayed deafferentation pain in the vicinity of the injured nerve. The related morbidity and challenges with pain recurrence restrict these irreversible procedures' long-term effectiveness [6]. Technological advances in neuromodulation have offered such refractory conditions a phenomenal perspective [7].

The trigeminal ganglion (TG) is a crucial element in the nociception of craniofacial pain and plays a role in the peripheral modulation of pain pathways in the TN [8]. Damaged TG neurons brought on by unintentional and spontaneous nerve lesions are known to induce severe pain and sensory abnormalities. TG had been thought to be a node of transition for sensory data commuting from the periphery to the center. The fact that it can function as an integrating structure in the peripheral nervous system to control intracellular modulation mechanisms, as well as intercellular and autocrine signaling, is now well documented [9]. Targeting this essential component of the trigeminal pain pathway has been increasingly popular in recent years. A thorough review is necessary to offer information on its potential.

In this review, PubMed and Scopus Index were used as major search engines to find relevant articles on trigeminal ganglion modulation approaches in trigeminal neuralgia. The preliminary search retrieved 34 non-duplicated entries from the database upon implementing the key terms without accounting for the publication date. Only 25 of them were significantly relevant to the subject and featured in this review. Additionally, we referenced literature from the Scopus index that was compiled for clarity concerning the TG and trigeminal pain circuitry.Here, we summarize the trigeminal ganglion-focused neuromodulation techniques employed to address trigeminal neuralgia. These studies demonstrate the significance of the trigeminal ganglion in TN, alterations to the TG following TN, multiple approaches for targeting the TG, and its prospects. As a potential therapeutic strategy for trigeminal neuralgia, we propose the targeted modulation of the trigeminal ganglion itself.

Characteristics of Trigeminal ganglion

The TG is a sensory ganglion that resides in the dura mater's Meckel cave. The ocular (V1), maxillary (V2), and mandibular (V3) nerves, which in rodents form a single thick bundle at their origin, are generated by the TG. Most of the peripheral axons of pseudo-unipolar primary afferent neurons are found in those three large cranial nerves [10]. The source of the central processes of the trigeminal afferents is the TG, which produces the trigeminal nerve and enters the brainstem at the pontine level. The TG is made up of glial cells and neurons[11]. The distinctive pseudounipolar architecture of TG neurons interjected links between the two terminals and the discharge of transmitters at both central and peripheral sites [12]. TG neurons can be categorized into two sizes: large and small. Based on established studies, the modulation of pain in TN is mediated by small-sized TG neurons that are mostly coupled to unmyelinated nerve fibers [13]. The glial cells that surround the pseudo-unipolar neurons have a substantial impact on the activity and function of the neurons by altering the ionic concentrations [14].

Neuropeptides and their receptors signaling in TG

TG expresses a wide range of neuropeptides. Due to their role in the trigeminal pain pathway, these neuropeptides are of great interest. Distinct neuropeptides secreted by TG are calcitonin gene-related peptide (CGRP), substance P (SP), and vasoactive intestinal peptide (VIP) [15]. Numerous additional neuropeptides have been outlined by immunostaining studies in TG, including neurokinin A, pituitary adenylate-cyclase-activating polypeptide receptor type 1 (PACAP), cholecystokinin, galanin, somatostatin, and opioid peptides. They contribute to signal transmission by serving as neuromodulators [16].

About 40–50% of the trigeminal system's neurons are CGRP-immunoreactive, making it the most dominant signaling neuropeptide in the region [17]. There are two isoforms of CGRP: α- and β-CGRP. Among them, α-CGRP predominates in sensory nerves with cell bodies in the dorsal root ganglion (DRG) and TG [18]. CGRP receptor expression in the TG is implicated in signaling pathways that could be crucial for sensitizing processes in the induction of headaches and facial pain [19]. TG neurons express transient receptor potential (TRP) family receptors in substantial concentrations. In most TG neurons, immunoreactivity for the TRP vanilloid type 1 receptor channel (TRPV1) has been found [20]. 10–30% of TG cells are SP-containing cells, which are dispersed throughout the region. TG neurons also have a high affinity for the SP receptor, neurokinin 1-receptor (NK1-R) [21]. VIP-positive cells make up 10–12% of TG neurons and are linked to TG cells that contain SP. A subpopulation of small-to medium-sized TG neurons contains PACAP, and members of the family of seven-transmembrane G protein-coupled receptors mediate its effects [22]. Only a small fraction of TG neurons are reactive to neuropeptide Y (NPY) as it is primarily produced in sympathetic neurons [23].

Orexins and oxytocin are additional neuropeptides that may participate as signals in the TG but are most likely derived from different sources. In rat TG neurons, orexin receptor (OX1R and OX2R) mRNA has been identified, of which the OX 2 R mRNA level was more abundantly expressed than the OX 1 R [24]. TG neurons have also been found to exhibit oxytocin receptor immunoreactivity [25].

Alterations in TG after trigeminal nerve injury

As per earlier research, the fundamental abnormality is centered in the TG rather than the skin or the central nervous system, and the sensation is substantially typical in between episodes of ectopic paroxysmal ganglion discharge [26]. Trigeminal nerve damage significantly alters the plasticity of TGs' biomarkers. Major alterations in injured TGs point to the possibility that NPY and Iba1 may be key players in the pathophysiology of orofacial neuropathic pain because of peripheral nerve injury [27]. The TG's parts V1, V2, and V3 are associated with each other, and stimulation of the V3 neurons may raise the amounts of active signaling proteins in the satellite glial cells (SGC) and neuronal cells in other ganglion regions, which aids in signal transmission and chronic pain. SGC undergoes modifications like an increase in gap junctions and the building of bridges that bind the perineuronal sheaths [28].

Following a trigeminal nerve injury, neuropeptides (such as SP and CGRP), several neurotrophic factors, glutamate, and ATP impact neuronal activity in both intact and injured TG neurons. Both the ganglion and the nerve are susceptible to these variations. The amplification of the perceptive neuronal excitability in the TG is facilitated by the active cell-to-cell interface among macrophages and TG neurons via various chemical messengers. As per reports, peripheral nerve damage triggers action potentials defined as injury discharges in the TG neurons, which are conveyed to the trigeminal spinal nucleus (Vc) [29].

Trigeminal ganglion stimulation approaches

Addressing the trigeminal nerve central terminal for trigeminal pain relief has been undertaken in a multitude of ways. It's an intriguing strategy to target the trigeminal ganglion itself. Below, we've explored various trigeminal ganglion stimulation approaches made to date for managing TN pain (Fig. 1 and Table 1).

Fig. 1
figure 1

Different trigeminal ganglion stimulation approaches for managing TN pain

Table 1 Trigeminal ganglion stimulation approaches in orofacial neuropathic pain

Electrical stimulation of TG

Few clinical studies have reported the analgesic effect in TN patients following electrostimulation of TG. The potential of TG stimulation to access multiple dermatomes with a single electrode renders it feasibly interesting. In a clinical trial study, it has been advised that patients who experience chronic trigeminopathic pain should always consider persistent therapeutic electrostimulation of the TG [33]. In certain studies, electrical stimulation of the trigeminal ganglion influences both the peripheral and the central terminals of the primary trigeminal sensory neuron. In response to the nociceptive input, it is postulated that the stimulation triggers the ascending nociceptive routes to the thalamus and the cortex, which then activate the descending modulatory system [30]. The TG, also known as the Gasserian ganglion, is a conjunction of all three trigeminal branches that permits one to designate precise facial areas based on the available somatotopic arrangement of nerve fibers while achieving a therapeutic effect on the full half of the face with minor surgical intervention. In refractory trigeminal neuropathy, stimulation of the Gasserian ganglion is an effective, minimally invasive, and non-destructive treatment that ought to be considered earlier in the treatment algorithm [31]. Despite a few risks, such as electrode dislocation (10–30%) and other mechanical defects (24%), TG electric stimulation has shown to be effective in treating patients with chronic trigeminal neuropathic pain and persistent idiopathic facial pain who have tried and failed or were not deemed candidates for standard surgical interventions [39].

Pharmacological modulation

Pharmacological approaches have also been made in TG to intervene in TN management. According to in vivo research by Minghan et al.,a Cdk5 inhibitor peptide, TFP5, can directly affect peripheral nerve pain signaling at the TG.In addition to reducing the polymodal nociceptor response to capsaicin and brush, direct treatment of TG with TFP5 significantly reduced integrated calcium signaling for both stimuli. Inhibition of Cdk5 reduces pain at the peripheral level via reducing pain signaling in peripheral TG neurons. Cdk5 activity at TG’s main sensory neurons could be the basis of more efficient and secure therapies for facial pain [40]. A different study used the organ culture method to investigate if botulinum neurotoxin A can directly interact with sensory processes in the trigeminal ganglion [41].

Another study revealed that oxytocin, a neuropeptide hormone, is crucial in mitigating TG neuronal hyperexcitability following nerve injury. The action is most likely attributed to vasopressin-1A receptor-mediated regulation of voltage-gated potassium channels [35]. Likewise, another group documented that the intraganglionic injection of oxytocin attenuated orofacial ectopic pain produced by inferior alveolar nerve transection. Targeting oxytocin receptors (OXTR) in TG may be a viable therapeutic strategy for treating chronic orofacial neuropathic pain since activation of OXTR in TG can diminish aberrant pain by inhibiting the action of CGRP, interleukin-1β, and tumor necrosis factor-α [34].

A clinical report also indicated that in patients with refractory TN, intra-arterial anesthetic injection to the TG could be employed to regulate trigeminal nerve activity. In the region of the middle meningeal artery close to the arterial branch serving the trigeminal nerve ganglion, researchers delivered intra-arterial lidocaine of up to 50 mg persession. The authors stated that consistent intraprocedural electrophysiologic suppression and short-term clinical improvement in subjects with resistant TN support the possibility of modulating trigeminal nerve activity via the arterial route [36].

Optogenetic modulation of TG

The technique of optogenetic modulation in TG is new and elusive in the literature. Not long ago, we published an article reporting that manipulating TG activity directly through an optogenetic approach might govern the varied attributes of TN pain sensations and elicit antinociception. We have demonstrated that thalamic relay nuclei and the trigeminal brainstem, which direct to the somatosensory cortex, are modified by halorhodopsin-mediated inhibition of TG. Additionally, we highlighted that the inhibition has a significant impact on bursting activity and that thehyperpolarization results in a general reduction in thalamic discharge [38].

DREADD activation of TG

Notably, Korczeniewska and the team approached the TG modulation with systemic administration of the DREADD agonist clozapine N-oxide (CNO). They claimed that by intervening in TG cells that are hyperactive, CNO-mediated activation of hM4Di-DREADDs in the TG improves nerve injury-induced neuropathic pain. According to the the authors, TG neuronal silencing resulted in significantly reduced behavioral responses to vibrissal pad mechanical stimulation and decreased c-fos expression in the brainstem trigeminal nucleus caudalis [37].

Other modulation approaches in TG

Apart from the approaches mentioned above, other modulation approaches include gamma knife radiosurgery (GKS), percutaneous radiofrequency (RF), and photobiomodulation (PBM) in TG. A study has postulated that given its short latency duration, low collateral risks, and a significant share of pain management, GKS on TG is indeed a feasible therapy choice. The inactivation of the satellite glial cells in the TG might well be associated with the action mechanisms of radiosurgery [42]. Radiosurgery in the TG induces a lesion to the SGC, which enables the degenerative mechanisms triggered by the affected neuron after a nerve injury to subside. An extracellular hyperpolarization that lasted longer could further result from reducing the overall SGCs' gaps and attributes. Clinically, pain discharges are inhibited and are sustained in this state for a considerable amount of time. Employing RF treatment of the TG for treating patients with TN has indicated high efficacy. However, there has also been a substantial level of adverse effects. Lower sensory stimulation was linked to more hypesthesia, whereas higher stimulation intensities were coupled with lower efficacy [43]. Another intriguing approach in TG was PBM, which centers on the dynamics of visible or near-infrared photons with the biomolecules present inside cells or tissues. In vitro studies revealed that effective PBM energy density, 1 and 2 J/cm2, and frequency, 10 Hz, promoted neuronal growth of TG neurons and might have therapeutic potential in circumstances prompting the regeneration of damaged TG neurons [44].

Discussion

The peripheral modulation of pain pathways in TN is facilitated by the trigeminal ganglion, a crucial element in the nociception of craniofacial pain. Trigeminal neuropathic pain syndromes, which are often less effectively treated with traditional surgical techniques like microvascular decompression and percutaneous rhizotomy, can be treated with TG stimulation, a neuromodulatory therapy. A study has reported that the trigeminal branch stimulation of the V3 dermatome carries a particularly significant risk of cutaneous erosion because of the lower jaw's extreme movement. Therefore, integrating TG stimulation, which also crosses the V3 distal nerve as it joins the ganglion, would be a feasible approach [32] (Fig. 2).

Fig. 2
figure 2

Synaptic connections of the trigeminal ganglion in the orofacial pain pathway. Three peripheral nerve branches (V1: ophthalmic; V2: Maxillary; V3; mandibular) of the trigeminal nerve, whose cell bodies sit in the trigeminal ganglion (TG), convey pain perception from the facial region and radiate centrally towards synapse with the second order neurons in the trigeminal spinal nucleus caudalis (Vc). Afterward, the second-order neurons ascend and end in the thalamus. Pain is mediated in the primary somatosensory cortex (S1), which receives nociceptive input from the thalamus. The maximized view of TG shows the prospects of targeting TG. Small-conductance calcium-activated potassium (SK) type 3 channel; Large conductance calcium-activated potassium (BKCa); γ-aminobutyric acid-A (GABAA) receptor; Satellite glial cells (SGC); VPM: Ventral posteromedial thalamus

Another possibility is targeting multiple TG channel pathways.Essential sodium channel barriers that are believed to occur in the dorsal root ganglion because of nerve injury also occur in the TG [45]. TN may indeed be managed with newer drugs targeting sodium channel subtypes in TG. Small-conductance calcium-activated potassium (SK) type 3 channel is present in TG and might be involved in TN pathogenesis. Thus, targeting this channel in TG may lay the basis for TN therapy [46]. It has been reported that TG expresses many large conductance calcium-activated potassium (BKCa) channels, which are thought to contribute to the anti-hyperalgesic action in neuropathic pain. To better comprehend mechanical allodynia, a study that focused on TG neurons discovered that the downregulation of BKCa channels in both injured and intact nociceptive TG neurons was a key factor in the TN [47]. They proposed that manipulating brain natriuretic peptide or its receptor or BKCa channel pathway in TG corresponds to a novel therapeutic point for the clinical management of TN [48]. Because TG contains γ-aminobutyric acid (GABA) and its receptors, a study investigated the masticatory muscle-evoked brainstem trigeminal neuron responses following intraganglionic injection of muscimol (GABAA) but not baclofen (GABAB). It was found that activating GABAA receptors might have a gating function on sensory transmission via TG by boosting innocuous sensory input and lowering putative nociceptive input [49].

In contrast to targeting TG in humans, there are some difficulties in targeting TG in rodents for preclinical studies. Only a few techniques fordelivering drugs or viral vectors to rodents' trigeminal ganglia have been described. Among them are the cranial and infraorbital routes. By targeting TG from the base of the skull, the dura mater's impact on the viral vector's diffusion into the trigeminal ganglia is avoided. As a result, it shielded the brain and peripheral nerves, limiting any likelihood of neuropathic pain after the injection. Additionally, it has been proposed that thetransduction of CGRP shRNA may partially reduce discomfort following TG injections [50]. We followed the stereotaxic technique to inject the optogenetic virus into TG as mentioned in earlier studies [51, 52]. In addition, we employed flexible optic fiber to modulate the TG activity upon stimulation, which ensures high-quality coupling and durability [38]. Specific targeting and understanding of the molecular perspectives in trigeminal ganglion may help highlight the potential therapeutic intervention in orofacial neuropathic pain.

Conclusion

Numerous brain areas have been investigated to date to modulate the trigeminal pain pathway. The trigeminal ganglion, a key component of the trigeminal pain circuitry, has also been studied using electrical, pharmacological, optogenetic, chemogenetic, radiological, and other methods. This review will help gain the new vision towards targeting TG directly for TN management.

Availability of data and materials

Not applicable.

Abbreviations

BKCa:

Large conductance calcium-activated potassium channels

CNO:

Clozapine N-oxide

CGRP:

Calcitonin gene-related peptide

Cdk5:

Cyclin-dependent kinase 5

DREADD:

Designer Receptors Exclusively Activated by Designer Drugs

DRG:

Dorsal root ganglion

GABAA:

γ-Aminobutyric acid A receptor

GFAP:

Glial fibrillary acidic protein

GKS:

Gamma knife radiosurgery

NpHR:

Halorhodopsin from Natronomonas

OXTR:

Oxytocin receptors

PACAP:

Pituitary adenylate-cyclase-activating polypeptide receptor type 1

PBM:

Photobiomodulation

SGC:

Satellite glial cells

SK3:

Small-conductance calcium-activated potassium type 3 channel

TG:

Trigeminal ganglion

TN:

Trigeminal neuralgia

TRP:

Transient receptor potential

TRPV1:

TRP vanilloid type 1 receptor channel

VIP:

Vasoactive intestinal peptide

VPm:

Ventral Posteromedial nucleus of the thalamus

References

  1. Bendtsen L, Zakrzewska JM, Heinskou TB et al (2020) Advances in diagnosis, classification, pathophysiology, and management of trigeminal neuralgia. Lancet Neurol 19:784–796. https://doi.org/10.1016/S1474-4422(20)30233-7

    Article  CAS  PubMed  Google Scholar 

  2. Cruccu G, di Stefano G, Truini A (2020) Trigeminal neuralgia. N Engl J Med 383:754–762. https://doi.org/10.1056/NEJMra1914484

    Article  PubMed  Google Scholar 

  3. Gambeta E, Chichorro JG, Zamponi GW (2020) Trigeminal neuralgia: an overview from pathophysiology to pharmacological treatments. Mol Pain 16:1744806920901890. https://doi.org/10.1177/1744806920901890

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Michael Silva & Aviv Ouanounou (2020) Trigeminal Neuralgia: Etiology, Diagnosis, and Treatment. SN Compr Clin Med 2:1585–1592. https://doi.org/10.1007/s42399-020-00415-9

  5. Lee CH, Jang HY, Won HS et al (2021) Epidemiology of trigeminal neuralgia: an electronic population health data study in Korea. Korean Journal of Pain 34:250–261. https://doi.org/10.3344/KJP.2021.34.3.332

    Article  Google Scholar 

  6. Hassanzadeh R, Jones JC, Ross EL (2014) Neuromodulation for intractable headaches. Curr Pain Headache Rep 18:1–8. https://doi.org/10.1007/s11916-013-0392-1

    Article  Google Scholar 

  7. Gupta K, Texakalidis P, Boulis NM (2021) Programming Parameters and Techniques in Trigeminal Ganglion Stimulation for Intractable Facial Pain. Neuromodulation: Technology at the Neural Interface 24:1100–1106. https://doi.org/10.1111/ner.13306

  8. Karl Messlinger AFR (2019) Current understanding of trigeminal ganglion structure and function in headache. Cephalalgia 39:1661–1674. https://doi.org/10.1177/0333102418786261

    Article  Google Scholar 

  9. Somaza S, Eglee M (2022) Novel theory about radiosurgery’s action mechanisms on trigeminal ganglion for idiopathic trigeminal neuralgia: role of the satellite glial cells. Surg Neurol Int 11:1–6

    Google Scholar 

  10. Lazarov NE (2002) Comparative analysis of the chemical neuroanatomy of the mammalian trigeminal ganglion and mesencephalic trigeminal nucleus. Elsevier

    Book  Google Scholar 

  11. Hanani M, Spray DC (2020) Emerging importance of satellite glia in nervous system function and dysfunction. Nat Rev Neurosci 21:485–498. https://doi.org/10.1038/s41583-020-0333-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Veríssimo CP, Filha LGA, da Silva FJM et al (2022) Short-term functional and morphological changes in the primary cultures of trigeminal ganglion cells. Curr Issues Mol Biol 44:1257–1272. https://doi.org/10.3390/cimb44030084

    Article  PubMed  PubMed Central  Google Scholar 

  13. Dasilva AF, Dossantos MF (2012) The role of sensory fiber demography in trigeminal and postherpetic neuralgias. J Dent Res 91:17–24. https://doi.org/10.1177/0022034511411300

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang L-YM, Yanping G (2011) Communication between neuronal somata and satellite glial cells in sensory ganglia. Glia 4:1571–1581. https://doi.org/10.1002/glia.22541.Communication

    Article  Google Scholar 

  15. Messlinger K, Balcziak LK, Russo AF (2020) Cross-talk signaling in the trigeminal ganglion: role of neuropeptides and other mediators. J Neural Transm 127:431–444. https://doi.org/10.1007/s00702-020-02161-7

    Article  PubMed  Google Scholar 

  16. Goto T, Iwai H, Kuramoto E, Yamanaka A (2017) Neuropeptides and ATP signaling in the trigeminal ganglion. Jpn Dent Sci Rev 53:117–124. https://doi.org/10.1016/j.jdsr.2017.01.003

    Article  PubMed  PubMed Central  Google Scholar 

  17. Iyengar S, Johnson KW, Ossipov MH, Aurora SK (2019) CGRP and the Trigeminal System in Migraine. Headache 59:659–681. https://doi.org/10.1111/head.13529

    Article  PubMed  PubMed Central  Google Scholar 

  18. Russell FA, King R, Smillie SJ et al (2014) Calcitonin gene-related peptide: physiology and pathophysiology. Physiol Rev 94:1099–1142. https://doi.org/10.1152/physrev.00034.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang Z, Winborn CS, de Prado BM, Russo AF (2007) Sensitization of calcitonin gene-related peptide receptors by receptor activity-modifying protein-1 in the trigeminal ganglion. J Neurosci 27:2693–2703. https://doi.org/10.1523/JNEUROSCI.4542-06.2007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dux M, Rosta J, Messlinger K (2020) TRP channels in the focus of trigeminal nociceptor sensitization contributing to primary headaches. Int J Mol Sci 21:342. https://doi.org/10.3390/ijms21010342

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gunjigake KK, Goto T, Nakao K et al (2006) Correlation between the appearance of neuropeptides in the rat trigeminal ganglion and reinnervation of the healing root socket after tooth extraction. Acta Histochem Cytochem 39:69–77. https://doi.org/10.1267/ahc.05057

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Edvinsson L, Tajti J, Szalárdy L, Vécsei L (2018) PACAP and its role in primary headaches. J Headache Pain 19(1):21. https://doi.org/10.1186/s10194-018-0852-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Oliveira MM, Akerman S, Tavares I, Goadsby PJ (2016) Neuropeptide y inhibits the trigeminovascular pathway through NPY Y1 receptor: implications for migraine. Pain 157:1666–1673. https://doi.org/10.1097/j.pain.0000000000000571

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cady RJ, Denson JE, Sullivan LQ, Durham PL (2014) Dual orexin receptor antagonist 12 inhibits expression of proteins in neurons and glia implicated in peripheral and central sensitization. Neuroscience 269:79–92. https://doi.org/10.1016/j.neuroscience.2014.03.043

    Article  CAS  PubMed  Google Scholar 

  25. Tzabazis A, Mechanic J, Miller J et al (2016) Oxytocin receptor: Expression in the trigeminal nociceptive system and potential role in the treatment of headache disorders. Cephalalgia 36:943–950. https://doi.org/10.1177/0333102415618615

    Article  PubMed  Google Scholar 

  26. Rappaport ZH, Devor M (1994) Trigeminal neuralgia: The role of self-sustaining discharge in the trigeminal ganglion. Pain 56:127–138. https://doi.org/10.1016/0304-3959(94)90086-8

    Article  PubMed  Google Scholar 

  27. Lynds R, Lyu C, Lyu GW et al (2017) Neuronal plasticity of trigeminal ganglia in mice following nerve injury. J Pain Res 10:349–357. https://doi.org/10.2147/JPR.S120092

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Costa FAL, Neto FLM (2015) Satellite glial cells in sensory ganglia: its role in pain. Brazilian Journal of Anesthesiology (English Edition) 65:73–81. https://doi.org/10.1016/j.bjane.2013.07.013

    Article  Google Scholar 

  29. Shinoda M, Kubo A, Hayashi Y, Iwata K (2019) Peripheral and central mechanisms of persistent orofacial pain. Front Neurosci 13:1–10. https://doi.org/10.3389/fnins.2019.01227

    Article  Google Scholar 

  30. Bohár Z, Fejes-Szabó A, Tar L et al (2013) Evaluation of c-Fos immunoreactivity in the rat brainstem nuclei relevant in migraine pathogenesis after electrical stimulation of the trigeminal ganglion. Neurol Sci 34:1597–1604. https://doi.org/10.1007/s10072-013-1292-1

    Article  PubMed  Google Scholar 

  31. Logghe Y, Smet I, Jerjir A et al (2021) Trigeminal neuropathy: two case reports of gasserian ganglion stimulation. Brain Behav 11:1–6. https://doi.org/10.1002/brb3.2379

    Article  Google Scholar 

  32. Gupta K (2022) Case Report: Novel Anchoring Technique and Surgical Nuances for Trigeminal Ganglion Stimulation in the Treatment of Post-Herpetic Trigeminal Neuropathic Facial Pain. Frontiers in Pain Research 3:1–6. https://doi.org/10.3389/fpain.2022.835471

    Article  Google Scholar 

  33. Mehrkens JH, Steude U (2007) Chronic electrostimulation of the trigeminal ganglion in trigeminal neuropathy: current state and future prospects. Acta Neurochir Suppl (Wien) 97:91–97. https://doi.org/10.1007/978-3-211-33081-4_11

    Article  CAS  Google Scholar 

  34. Huang CL, Liu F, Zhang YY et al (2021) Activation of oxytocin receptor in the trigeminal ganglion attenuates orofacial ectopic pain attributed to inferior alveolar nerve injury. J Neurophysiol 125:223–231. https://doi.org/10.1152/JN.00646.2020

    Article  CAS  PubMed  Google Scholar 

  35. Kubo A, Shinoda M, Katagiri A et al (2017) Oxytocin alleviates orofacial mechanical hypersensitivity associated with infraorbital nerve injury through vasopressin-1A receptors of the rat trigeminal ganglia. Pain 158:649–659. https://doi.org/10.1097/j.pain.0000000000000808

    Article  CAS  PubMed  Google Scholar 

  36. Qureshi AI, Saleem MA, Jadhav V et al (2018) Intra-arterial Modulation of the Trigeminal Nerve Ganglion in Patients with Refractory Trigeminal Neuralgia. J Neuroimaging 28:79–85. https://doi.org/10.1111/jon.12476

    Article  PubMed  Google Scholar 

  37. Korczeniewska OA, James MH, Eliav T et al (2022) Chemogenetic inhibition of trigeminal ganglion neurons attenuates behavioural and neural pain responses in a model of trigeminal neuropathic pain. Eur J Pain (United Kingdom) 26:634–647

    CAS  Google Scholar 

  38. Kc E, Islam J, Kim S et al (2022) Pain Relief in a Trigeminal Neuralgia Model via Optogenetic Inhibition on Trigeminal Ganglion Itself With Flexible Optic Fiber Cannula. Front Cell Neurosci 16:1–14. https://doi.org/10.3389/fncel.2022.880369

    Article  CAS  Google Scholar 

  39. Kustermans L, van Buyten JP, Smet I et al (2017) Stimulation of the Gasserian ganglion in the treatment of refractory trigeminal neuropathy. Journal of Cranio-Maxillofacial Surgery 45:39–46. https://doi.org/10.1016/j.jcms.2016.10.014

    Article  PubMed  Google Scholar 

  40. Hu M, Doyle AD, Yamada KM, Kulkarni AB (2022) Visualization of trigeminal ganglion sensory neuronal signaling regulated by Cdk5. Cell Rep 38:110458. https://doi.org/10.1016/j.celrep.2022.110458

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Edvinsson J, Warfvinge K, Edvinsson L (2015) Modulation of inflammatory mediators in the trigeminal ganglion by botulinum neurotoxin type A: an organ culture study. J Headache Pain 16:555. https://doi.org/10.1186/s10194-015-0555-z

    Article  CAS  PubMed  Google Scholar 

  42. Somaza S, Montilla EM, Mora MC (2019) Gamma knife radiosurgery on the trigeminal ganglion for idiopathic trigeminal neuralgia: results and review of the literature. Surg Neurol Int 10:1–9. https://doi.org/10.25259/SNI-134-2019

    Article  Google Scholar 

  43. M Koning v., Koning NJ, Koning HM, van Kleef M, 2014 Relationship between sensory stimulation and side effects in percutaneous radiofrequency treatment of the trigeminal ganglion Pain Pract 14 581 587 https://doi.org/10.1111/papr.12124

  44. Cho H, Jeon HJ, Park S et al (2020) Neurite growth of trigeminal ganglion neurons in vitro with near-infrared light irradiation. J Photochem Photobiol B 210:111959. https://doi.org/10.1016/j.jphotobiol.2020.111959

    Article  CAS  PubMed  Google Scholar 

  45. Eriksson J, Jablonski A, Persson AK et al (2005) Behavioral changes and trigeminal ganglion sodium channel regulation in an orofacial neuropathic pain model. Pain 119:82–94. https://doi.org/10.1016/j.pain.2005.09.019

    Article  CAS  PubMed  Google Scholar 

  46. Zhao J, Zhang Y, Liu X et al (2021) Activation of SK3 channel plays a pivotal role in modulation of trigeminal neuralgia. Neurol Res 43:1005–1012. https://doi.org/10.1080/01616412.2021.1948765

    Article  CAS  PubMed  Google Scholar 

  47. Liu CY, Lu ZY, Li N et al (2015) The role of large-conductance, calcium-activated potassium channels in a rat model of trigeminal neuropathic pain. Cephalalgia 35:16–35. https://doi.org/10.1177/0333102414534083

    Article  PubMed  Google Scholar 

  48. Xu W, Yao Y, Zhu D et al (2021) Involvement of the BNP/NPR-A/BKCa pathway in rat trigeminal ganglia following chronic constriction injury. J Neurophysiol 125:1139–1145. https://doi.org/10.1152/jn.00682.2020

    Article  CAS  PubMed  Google Scholar 

  49. Ranjbar Ekbatan M, Cairns BE (2021) Attenuation of sensory transmission through the rat Trigeminal Ganglion by GABA Receptor Activation. Neuroscience 471:80–92. https://doi.org/10.1016/j.neuroscience.2021.07.018

    Article  CAS  PubMed  Google Scholar 

  50. Long H, Liao L, Zhou Y et al (2017) A novel technique of delivering viral vectors to trigeminal ganglia in rats. Eur J Oral Sci 125:1–7. https://doi.org/10.1111/eos.12326

    Article  CAS  PubMed  Google Scholar 

  51. Whitehead JL, Ohara PT, Tauscher AN, LaVail JH (2003) A procedure to deliver herpes simplex virus to the murine trigeminal ganglion. Brain Res Protoc 12:60–66. https://doi.org/10.1016/S1385-299X(03)00072-2

    Article  Google Scholar 

  52. XF Tang eng, Bi RY un, Meng Z, Gan YH ua, 2014 Stereotaxis of mandibular nerve initial point of trigeminal ganglion in rats Chinese Journal of Dental Research 17 99 104 https://doi.org/10.3290/j.cjdr.a33272

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Research Foundation of Korea (NRF2020R1F1A1052716); the Brain Korea 21 FOUR of the National Research Foundation of Korea (NRF) funded by the Ministry of Education (No. 5199990614277). This study was additionally supported by the "Regional Innovation Strategy (2020RIS0188, 2021RIS0096)" through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (MOE). This work was also supported by Chungbuk National University BK21 program (2022).

Author information

Authors and Affiliations

Authors

Contributions

EK and YSP contributed to the manuscript’s conceptualization and design. EK drafted the initial manuscript and prepared the figures. All authors contributed to the critical revision of this paper and approved the final version. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Young Seok Park.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors approved the publication of this paper.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

KC, E., Islam, J. & Park, Y.S. Trigeminal ganglion itself can be a viable target to manage trigeminal neuralgia. J Headache Pain 23, 150 (2022). https://doi.org/10.1186/s10194-022-01512-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s10194-022-01512-x

Keywords