Next Article in Journal
Carrot DcALFIN4 and DcALFIN7 Transcription Factors Boost Carotenoid Levels and Participate Differentially in Salt Stress Tolerance When Expressed in Arabidopsis thaliana and Actinidia deliciosa
Previous Article in Journal
Integrated Bioinformatic Investigation of EXOSCs in Hepatocellular Carcinoma Followed by the Preliminary Validation of EXOSC5 in Cell Proliferation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pain Management and Rehabilitation for Central Sensitization in Temporomandibular Disorders: A Comprehensive Review

1
Dentistry Unit, Department of Health Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
2
Physical Medicine and Rehabilitation Unit, Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
3
Institute of Neurology, Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
4
Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
5
Neurological Rehabilitation Unit, Istituti Clinici Scientifici Maugeri, IRCCS Institute of Bari, 70124 Bari, Italy
6
Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12164; https://doi.org/10.3390/ijms232012164
Submission received: 29 September 2022 / Revised: 7 October 2022 / Accepted: 9 October 2022 / Published: 12 October 2022
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Temporomandibular disorders (TMD) are a group of musculoskeletal diseases affecting masticatory muscles and temporomandibular joints (TMJ). In this context, the chronic TMD could be considered as a condition with chronic primary orofacial pain, presenting as myofascial TMD pain or TMJ arthralgia. In this context, myogenous TMD may present overlapping features with other disorders, such as fibromyalgia and primary headaches, characterized by chronic primary pain related to dysfunction of the central nervous system (CNS), probably through the central sensitization. This phenomenon could be defined as an amplified response of the CNS to sensory stimuli and peripheral nociceptive, characterized by hyperexcitability in the dorsal horn neurons in the spinal cord, which ascend through the spinothalamic tract. The main objectives of the management of TMD patients are: decreasing pain, increasing TMJ function, and reducing the reflex masticatory muscle spasm/pain. The first-line treatments are physical therapy, pharmacological drugs, occlusal splints, laser therapy, extracorporeal shockwave therapy, transcutaneous electrical nerve stimulation, and oxygen–ozone therapy. Although all these therapeutic approaches were shown to have a positive impact on the central sensitization of TMD pain, there is still no agreement on this topic in the scientific literature. Thus, in this comprehensive review, we aimed at evaluating the evidence on pain management and rehabilitation for the central sensitization in TMD patients.

1. Introduction

Temporomandibular disorders (TMD) are a group of musculoskeletal and neuromuscular conditions affecting the masticatory muscles, the temporomandibular joint (TMJ), and the other associated structures [1]. According to the Diagnostic Criteria for TMD (DC/TMD) Axis I, TMD could be divided in intra-articular disorders, including disc displacement, arthralgia, arthritis, and arthrosis, and muscle disorders [1]. These latter are also defined as “myogenous TMD”, which can be further categorized into: local myalgia, if the pain is localized during palpation; myofascial pain, if the pain spreads within the palpated muscular territory; and myofascial pain with referral, if the pain spreads beyond the boundary of the masticatory muscles [1]. A recent systematic review and meta-analysis, with a combined sample of 2518 subjects, suggested that the prevalence of TMD could range from 25.2% to 34.9%, with a predominance of myofascial pain diagnosis (10.3–15.4%) [2]. The etiology is not clear, and it has been accepted as multifactorial, considering the multitude of initiating, predisposing, or perpetuating risk factors, including postural and parafunctional habits, repetitive microtrauma, direct and indirect trauma, and psychological factors, such as depression and anxiety [3]. The persistent and recurrent pain generated by the myofascial pain may cause limitations in the main activities of daily living (ADLs) and reduce the oral health-related quality of life (OHRQoL) [4]. If odontogenic causes are excluded [5,6], the painful TMD could be considered as the main cause of pain in the orofacial region [2,4].
More in detail, according to the 11th version of the International Classification of Diseases (ICD-11), the chronic primary pain was described as “pain in one or more anatomical regions that persists or recurs for longer than 3 months; is associated with significant emotional distress and/or significant functional disability; and the symptoms are not better accounted by another diagnosis” [7,8]. In this context, chronic TMD could be considered as conditions with chronic primary orofacial pain, presenting as myofascial TMD pain or TMJ arthralgia [7,8]. Compared to arthrogenous TMD, which appears to be a localized phenomenon, myogenous TMD may present overlapping features with other disorders, such as fibromyalgia and primary headaches, characterized by chronic primary pain related to dysfunction of the central nervous system (CNS), probably through the phenomenon of central sensitization [9,10,11,12,13].
Central sensitization can be defined as an amplified response of the CNS to sensory stimuli and peripheral nociceptive, characterized by hyperexcitability in the dorsal horn neurons in the spinal cord, which ascend through the spinothalamic tract [14,15,16,17]. Central sensitization could lead to the development of increased pain sensation from noxious stimuli, known as hypersensitivity, or pain originating from non-noxious stimuli, known as allodynia [14]. Moreover, other clinical features of central sensitization could be: increased temporal pain, summation spontaneous pain, referred pain, and pressure hyperalgesia [14,18].
Thus, the central sensitization could represent the basis of chronic pain “or pain that persists beyond a normal time of healing” in patients affected by TMD [19,20]. In the scientific literature, the central sensitization showed to have a role not only in the pathophysiology of TMD but also in other several chronic pain conditions, including: fibromyalgia, migraine, tension-type headache, irritable bowel syndrome, and chronic fatigue syndrome [21,22,23].
The psychological component in terms of emotional distress should be considered during the diagnosis process to better manage chronic pain conditions [8]. Anxiety, frustration, and depression may contribute to the development and to the persistence and exacerbation of pain [7,8].
Concerning the management of patients with TMD, it should be taken into consideration that the main objectives are: decreasing pain, increasing TMJ function, and reducing reflex masticatory muscle spasm/pain [24]. The first-line treatment for TMD is considered the conservative approach [25,26], including physical therapy [27], biofeedback [28], pharmacological drugs [29], TMJ injections [30], occlusal splints [31,32], laser therapy [33], extracorporeal shockwave therapy (ESWT) [34], transcutaneous electrical nerve stimulation (TENS) [35], and oxygen–ozone therapy [36].
All these therapeutic approaches might have a positive impact on the central sensitization of TMD pain (see Figure 1), albeit there is still no agreement in the scientific literature on this topic in terms of the management of these peculiar patients.
It should be noted that myogenous TMD might present a chronic primary pain related to a CNS dysfunction due to the mechanism of central sensitization that could lead to hypersensitivity in TMD patients [14]. To date, the scientific literature still lacks strong evidence on the key role of central sensitization for chronic pain in TMD patients affected by TMD [19,20], probably because this phenomenon showed to have a role in the pathophysiology of other chronic diseases [21,22,23]. Therefore, the diagnosis and the treatment of the central sensitization should be better investigated, taking into account the positive results that some conservative approaches might have on the central sensitization of TMD pain [25,26,27,28,29,30,31,32,33,34,35,36].
In this context, by the present comprehensive review, we aimed to investigate the state-of-the-art therapies regarding pain management and rehabilitation for the central sensitization in TMD patients in order to adequately manage this detrimental condition.

2. Central Sensitization

In a neurophysiological scenario, two main physiological phenomena, namely central sensitization and impairment of the inhibitory system of descending pain, together with the physiological mechanism of neuronal convergence, might be considered fundamental factors explaining TMD clinical patterns of and the associations with other comorbidities [15,24]. Other important mechanisms, such as central facilitation, peripheral sensitization, and neuroimmune alterations, also contribute to the physiological frame and rationale underlying the coexistence of TMD and painful conditions [15,24]. Furthermore, the nociceptive pathways responsible for TMD and headache are similar; in fact, there is an important area in the association between these disorders: the spinal trigeminalis nucleus—in particular, the caudal subnucleus [37]. This region is primarily responsible for nociceptive input from the head and face; therefore, it could be considered the first “meeting point” between TMD and headache disorders [38]. The generic term central sensitization is all neuronal alterations that may follow nociceptive processing within the caudal subnucleus in patients with chronic trigeminal pain. In short, this phenomenon may refer to a group of changes in the arrangement and quantity of membrane channels and neurotransmitters that ultimately decrease neuronal threshold activation, increase the firing rate, and widen receptor fields [39].
Many mechanisms are involved in central sensitization, but two main events (i.e., activation of the N-methyl-D-aspartate receptor and inhibition of gamma aminobutyric acid (GABA) and glycine receptors) are assumed to be present in most chronic pain conditions [40,41]. Furthermore, additional neurotransmitters, e.g., substance P (SP) and the calcitonin gene-related peptide, both released by small fiber neurons, relate to the lasting depolarization of the neuronal membrane and the sum of nociceptive inputs [42]. In addition to this hyperexcitable plasticity of the central nervous system, some mechanisms include basic inhibitory activity and, consequently, facilitate nociceptive signaling. Inhibition of the activity of GABAergic and glycinergic interneurons is assumed to reduce this second inhibitory activity, which, in turn, can increase the depolarization and excitation of order neurons [43]. Positive feedback circuits can amplify these large, second-order central changes: dynamic range neurons. Finally, this state of hyperexcitability could be a gradual and frequency-dependent facilitation of the nociceptive (wind-up phenomena) states of chronic pain [44].

3. Diagnosis of Central Sensitization in TMD

The diagnosis of the central sensitization is mandatory for adequate management, and, at the same time, it is considered a challenge that should be overcome in the near future. The central sensitization is an important aspect that is involved in the pathophysiology of different musculoskeletal painful conditions, including TMD [45]. Although the central sensitization could not be directly measured, different assessment tools have been developed to measure the sensory experiences that are greater than expected in amplitude, duration, or spatial extent [46].

3.1. Algometry and Pressure Pain Thresholds

In this scenario, a pressure algometer could be an effective tool in the screening and evaluation of patients with muscle pain due to central sensitization [47]. Pressure algometry is predominantly a manual procedure that requires a perceptual response from the participant or patient and is commonly used in quantitative sensory testing (QST), used extensively in clinical and experimental pain studies [48]. More in detail, the pressure (i.e., force per area often expressed in kPa) that the participants first perceive to be painful is defined as the pain threshold, and the maximum pressure endured by the participants is defined as the tolerance threshold.
In this context, the pressure pain threshold (PPT), defined as the minimum amount of pressure capable of inducing pain, is also frequently used in the evaluation of hyperalgesia [49]. The reliability of PPT depends not only on the application technique of the observer but also on the ability of the patient or participant to provide a consistent verbal indication of the PPT level [50]. The simplest algometry type allows the assessment of pressure using pressure-sensitive devices fitted to the finger [51]. Hand-held devices based on spring coil systems are also frequently used [52]. More sophisticated pressure algometers commonly provide visual feedback on the pressure application rate, which has been shown to influence the pain threshold [53]. In this scenario, it was demonstrated that patients with dysfunctions of the masticatory system showed variations in the repeatability of the pain threshold during pressure algometry [54]. More specifically, in TMD patients, the algometer can be applied to the masseter and temporal muscles and to the lateral pole of the condyle, recording the measurement in Kg when the patient felt pain or placing the algometer to measure 1 cm2 in direct contact with the joint and applied increasing pressure until the patient reported pain [55,56].

3.2. Assessment of Orofacial Somatosensory Function

The assessment of somatosensory function within the distribution of pain plays a central role in the certainty of the TMD diagnosis [57,58,59]. In this context, the QST is a psychophysical test procedure used to quantify the functional state of the somatosensory system of a patient by means of calibrated, graded innocuous or noxious stimuli and subjective perception thresholds [60]. Furthermore, QST comprises a battery of somatosensory tests assessing the response to a variety of standardized noxious and innocuous stimuli in affected and neighboring regions [61]. The feasibility of adapting the protocol to the orofacial region has been demonstrated recently [62]. Specifically, it has been shown that all 13 somatosensory tests can be performed on the apex of the tongue and facial gingiva in the upper jaw with moderate to excellent reliability for most measures. The duration of the intraoral examination per test site is in the range of 35 min, which is a bit slower than on extraoral sites [57].

3.3. Temporomandibular Disorders, Primary Headaches, and Cervical Pain

Painful orofacial and neck comorbidities are often associated with the TMD [63]. These coexisting conditions (particularly, headache, migraine, and neck pain) are not only highly associated with chronic pain-related TMDs but also increase the risk of their development [38,64,65]. The International Classification of Headache Disorders (ICHD) [66] and the DC/TMD [1] consider the main characteristics of pain in headaches and TMD, respectively. There are several hypotheses attempting to explain the association between TMD and headaches, including neuronal convergence, central sensitization, and inhibition of the descending pain downregulation mechanisms [67,68]. The strict relationship between TMD, headaches, and neck pain has been recently evaluated, not only in terms of sharing common pathogenic mechanisms and clinical features but also considering that one condition might influence or promote the development of another [11,64,69]. These conditions can cause facial pain and are frequently associated with the development of craniofacial allodynia during painful exacerbation [12]. Indeed, pain in both conditions has been attributed to common dysfunctions of the central pain regulation mechanisms [70,71]. On the other hand, the concomitance of TMD and migraines has shown worse levels of hyperalgesia and cutaneous allodynia, probably due to the sensitization of the central and peripheral nervous systems and the impairment of the descending modulatory pain pathways [44,70].

3.4. Evaluation Tools for Central Sensitization

The central sensitization should be adequately assessed, and one of the main evaluation scales in this field is the Central Sensitization Inventory (CSI) [72]. It includes two parts: part A, comprising 25 symptoms, such as physical symptoms, emotional distress, headache/jaw symptoms, and urological symptoms, and part B, consisting of 10 diseases, to evaluate central sensitization syndrome (CSS) [72]. A 40-point cut-off score of CSI part A was recommended to classify the presence of CSS in patients with chronic pain [73]. CSI development was based on a group of chronic pain conditions that has, probably among other mechanisms, central sensitization as a common putative pathophysiological mechanism [22]. The CSI has excellent test–retest reliability and internal consistency; the clinical and experimental characteristics of CSI could be commonly observed across many different chronic pain conditions, such as pelvic pain, osteoarthritis, spinal pain, and hereditary neuropathy [74,75,76,77]. Another evaluation tool is the Pain Sensitivity Questionnaire (PSQ), developed to assess various aspects of the clinical pain perceived [78]. The PSQ is a 17-item questionnaire that assesses a patient’s perceptions to various imagined physical stimuli that may be experienced in daily life [79]. Participants are asked to rate the pain intensity of each situational item on a 11-point scale, with 0 meaning “not painful at all” and 10 meaning “worst pain imaginable”. Three items (items 5, 9, and 13) are not normally rated as painful and are not included in the scoring. The PSQ-total score is the average of all items, except for the three nonpainful items. The PSQ-minor score is the average of items 3, 6, 7, 10, 11, 12, and 14—items, on average, that are perceived as causing minor pain. In patients with chronic localized pain, the PSQ-total and PSQ-minor scores have shown stronger correlations to QST compared to those found in healthy individuals [80].
Lastly, the Sensory Hypersensitivity Scale (SHS) is a 25-item instrumental tool assessing general and specific sensitivity and appears to be suitable as a screening instrument for central sensitization [81,82], albeit further studies should be performed to determine it in patients with TMD.

4. Evidence for Central Sensitization in TMD

To date, the scientific evidence on central sensitization in TMD in terms of diagnosis and treatment is still lacking.
In 2018, La Touche et al. [83] carried out a systematic review with a meta-analysis on central sensitization in TMD patients to summarize the scientific knowledge on this topic. The 22 included studies assessed the mechanical hyperalgesia (pressure pain thresholds), the thermal hyperalgesia (hot and cold pain thresholds), and the central hyperexcitability. Twelve studies evaluated the PPT, and eight of them revealed that PPT was significantly lower in TMD patients when compared to the control. Moreover, the meta-analysis showed strong evidence in favor of greater trigeminal pain sensitivity pressure in these patients. However, regarding thermal hyperalgesia, the meta-analysis comparing local and remote pain thresholds in patients with TMD and asymptomatic controls indicated that there were no significant differences in both cold and heat pain sensitivity between the groups. Lastly, spinal and central hyperexcitability were reported in TMD patients, as exhibited by increased mechanical temporal summation. Thus, the authors concluded that the findings of their systematic review and meta-analysis suggested the presence of peripheral and central nervous system sensitization in TMD patients.
However, the authors did not distinguish among different TMD subtypes, which are known to share different etiologies and pain mechanisms. Therefore, in 2021, Meng et al. [84] performed a meta-analysis to evaluate evidence in patients affected by only muscle pain-related TMD according to the DC/TMD [1]. The results provided evidence that, compared with the controls, these patients had reduced PPT and mechanical pain thresholds, whereas no evidence of changes in the cold detection threshold, warm detection threshold, heat pain threshold, cold pain threshold, and mechanical detection threshold were found.
Fernandez-de-las-Penas et al. [85] investigated bilateral, widespread pressure–pain hypersensitivity in nerve, muscle, and joint tissues in myofascial TMD women and controls. Their results showed significant differences between groups, but not between sides, for PPT levels over the supraorbital, infraorbital, mental, median, radial, and ulnar nerves; over the lateral pole of the TMJ; and over the tibialis anterior muscle. Thus, the results suggested both trigeminal and extratrigeminal sensitization of afferent inputs from neural tissues in myofascial TMD. An explanation could be related to the antidromic discharges originating from the central nervous system that may cause sensitization of peripheral nerve trunks that may depolarize nociceptive second-order neurons [85,86].
Additionally, in the scientific literature, it was shown that myofascial TMD patients presented larger referred-pain areas after intramuscular injection of hypertonic saline into the masseter muscle [87] and greater temporal summation of pain [88].
Furthermore, it should be noted that therapeutic strategies applied for managing central sensitization in TMD pain might be grouped into: bottom-up (i.e., tissue-based impairment treatments) and top-down interventions (i.e., strategies targeting the central nervous system) [89]. In this scenario, the bottom-up strategies could consist of joint-, soft tissue-, and nerve-targeting interventions, whereas top-down strategies might include physical therapy, motor imagery, and pain neuroscience education [89]. Therefore, we could conclude that, to date, the scientific literature showed that multimodal approaches seem to be more effective in patients with TMD, albeit it should be considered that the presence of depression might definitely increase pain sensitivity [90].
In this context, COVID-19 distress might have a negative impact on their psychological status, features of central sensitization, and facial pain severity in TMD patients [91].

5. Pharmacological Therapy and Central Sensitization in TMD

The purpose of pharmacological treatment of TMD is to alleviate the craniofacial pain linked to this condition, which is usually the main reason why these patients request medical attention [92,93]. Several therapeutic trials for TMD considered pain as the main outcome, evaluated with scales such as the visual analogic scale (VAS).
The mechanisms regarding chronic craniofacial pain remain to be fully elucidated [94]. Preclinical studies suggest that changes in both peripheral inputs and central brain structures could initiate and sustain chronic pain in TMD [94,95,96,97]. Inflammation with the release of many chemical mediators in TMD could result in the activation or peripheral sensitization of nociceptive endings of the trigeminal nerve [94]. Several mediators are linked to the peripheral sensitization, such as gamma amino butyric acid (GABA), serotonin, glutamate, and neuropeptides [94,95]. Conversely, the central sensitization strictly depends on an unbalance in the inhibitory and facilitatory descending pain modulatory systems, which promote and contribute to sustained chronic TMD pain [96]. Increased knowledge of those mechanisms underlying peripheral and central sensitization could improve pain management for TMD patients.
Pharmacological treatment for TMD pain could be very challenging for several reasons. First, as indicated above, multiple central and peripheral not yet fully elucidated mechanisms are involved in craniofacial TMD pain. Second, TMD pain is often associated with special emotional and psychological meanings, so a multidisciplinary approach could be indicated. Third, randomized controlled trials for the pharmacotherapy of these specific conditions are still lacking [98], so treatment is usually empirical. A Cochrane review evaluating TMD medications only included 11 studies in the qualitative synthesis [98]. The authors found lacking evidence to support or refute the efficacy of any drug for the treatment of TMD pain [98]. However, several medications typically prescribed in TMD have proven their effects for other craniofacial conditions.
The following subsections describe the most used drugs for the management of this condition, which are: nonsteroidal anti-inflammatory drugs (NSAIDs), beta-blockers, antidepressants, anti-seizure medications (ASMs), and opioids, along with other therapeutic approaches and new perspectives.

5.1. Nonsteroidal Anti-Inflammatory Drugs (NSAIDs)

NSAIDs are the most common class of medications prescribed for the treatment of craniofacial pain and have a proven efficacy for relief pain in TMD [99]. This class of medications include several molecules that exert their action inhibiting cyclo-oxygenase, thereby preventing the formation of prostaglandins. These drugs are usually well-tolerated and should be administrated for a minimum of two weeks to achieve an anti-inflammatory effect in TMD [100]. The main disadvantages of NSAID treatment could be: gastrointestinal adverse events, exacerbation of hypertension, and interactions with multiple drugs. The efficacy of topical or oral NSAIDs for the management of TMD pain is supported by controlled studies [101]. Ta and Dionne performed a six-week randomized double-blinded controlled trial comparing the efficacy of celecoxib, naproxen, and placebo for the treatment of TMD pain [101]. The authors showed that naproxen (500 mg twice a day) significantly reduced the clinical symptoms of TMD compared with celecoxib or a placebo [101]. De Carli and colleagues, in a double-blinded, randomized trial, showed that piroxicam, a cyclo-oxygenase-2 inhibitor, exhibited the lowest pain at a 30-day follow-up compared with the placebo [102]. Furthermore, Businco et al. showed that both topical and oral diclofenac are equally effective in the treatment of temporomandibular joint dysfunction symptoms [103]. One of the main advantages of the topical administration of diclofenac is preventing systemic adverse events of NSAIDs [103]. Topical creams of NSAIDs such as diclofenac could also reduce pain through the peripheral NMDA receptor antagonism [104]. Recently, a systematic review, including 11 randomized trails evaluating NSAIDs for the management of TMD, supported the use of NSAIDs in patients with TMD for the relief of pain [105].

5.2. Beta-Blockers

Beta-blockers exert their action by beta-adrenergic receptor antagonists. These drugs are widely used in other craniofacial conditions, such as for migraine prophylaxis [106]. Propranolol, a nonselective beta-blocker, is one of the most effective first-line drugs used for migraine prophylaxis [106]. The rationale for using beta-blockers to manage TMD pain comes from preclinical and animal models [107,108]. It was demonstrated that the activation of β₁ and β₂ adrenoceptors located in the TMJ region promotes serotonin (5-HT)-induced nociception [107,108]. To date, only few randomized controlled studies [98,99,100,101,102,103,104,105,106,107,108,109] have evaluated beta-blockers for TMD pain relief. A multicenter placebo-controlled trial using a “facial pain index” (FPI) as the primary endpoint evaluated the efficacy of propranolol in 200 TMD patients [109]. The authors showed that propranolol 60 mg twice a day was efficacious in achieving ≥30% and ≥50% FPI reductions after 9 weeks of treatment [109]. Tchivileva and colleagues showed a greater effect of propranolol in reducing FPI in migraineur TMD patients, suggesting that beta-blockers could be a reasonable option for treating the comorbidities of TMD and migraine [109].

5.3. Antidepressants

Both tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors (SSRIs) have been reported to reduce pain in TMD patients. These molecules act by binding 5HT receptors, thus producing a significant modulation of the nociceptive system. TCAs, particularly amitriptyline and nortriptyline, have been extensively used for both the prevention of primary headaches and for the treatment of myofascial masticatory chronic pain [63,110,111]. Among various antidepressants, TCAs and SSRIs seem to be the most effective for chronic orofacial pain [112]. A placebo-controlled study with a 14-day follow-up demonstrated a significant reduction in pain and discomfort of TMD patients treated with 25 mg/day of amitriptyline compared with the placebo [113]. A systematic review suggests a type B level of recommendation in favor of using TCAs for TMD [114]. Among SSRIs, paroxetine, duloxetine, and citalopram have been used for treating TMD symptoms [115]. A recent study suggested a better outcome for TMD management when including a combination of duloxetine 30 mg twice daily and TMJ arthrocentesis [115]. Thus, we might conclude that doses of antidepressant drugs, if used to treat TMD pain, should be lower than those used to control depression symptoms.

5.4. Antiseizure Medications

Antiseizure medications (ASMs) were extensively used to treat neuropathic pain and primary headaches such as migraines. These drugs act at several sites of action, reducing neural hyperexcitability. The relevant sites of action include both voltage- and ligand-gated ion channels. Among several ASMs, gabapentin and pregabalin were extensively used for managing chronic facial pain [29,116]. These compounds were structurally related to GABA, the main inhibitory neurotransmitter in the central nervous system; in this context, ASMs could be considered as an alternative therapy for refractory TMDs [29]. Kimos et al. [116] randomized 44 patients with TMD pain either to take gabapentin or a placebo, demonstrating that gabapentin had a statistically significant effect over the placebo in reducing spontaneous pain in the TMJ and the number of tender sites on the muscles of mastication. Some ASMs such as clonazepam or diazepam belong to the class of benzodiazepines, which enhance the response to GABA by facilitating the opening of chloride channels and, thus, cause hyperpolarization. BDZ have an antiseizure, anxiolytic, muscle relaxant, and hypnotic effect; thus, they can modulate TMD pain at several levels. Harkins et al. conducted a 60-day double blinded, randomized trial, comparing clonazepam versus a placebo in the management of chronic TMD pain [117]. As argued by a Cochrane meta-analysis [98], clonazepam did not show a statistically significant difference compared to a placebo on pain in the right or left temporomandibular joints [117]. Another double-blinded clinical trial, in which patients with TMD pain were randomized to take clonazepam, cyclobenzaprine, or a placebo, failed to demonstrate statistically significant differences between clonazepam and cyclobenzaprine compared to the placebo on jaw pain [118].

5.5. Opioids

Opioids are a class of analgesic drugs, which act at the central and peripheral opioid receptors, resulting in the blockage of painful neural inputs. It has also been postulated on the existence of a peripheral m-opioid receptor event at the level of the TM joint, thus providing a rationale for their topical use in the management of TMD pain [119,120]. The most common prescribed opioids for oral administration are codeine and oxycodone, but their use is not recommended. If prescribed, they should be used for a short period only in patients complaining about severe TMD pain, refractory to other treatments [121].

5.6. Other Therapies and New Perspectives

Many other types of drugs are used for the management of TMD pain, such as corticosteroids or muscle relaxants such as cyclobenzaprine [122]. A meta-analysis showed that cyclobenzaprine could ameliorate TMD muscle pain in the short term through its effect on local spasms and the associated acute pain [123].
As demonstrated above, multiple oral drugs are quite effective in the management of TMD pain; however, systemic adverse events for oral drugs raise issues for any long-term treatment strategy. Future perspectives include novel delivery systems for therapeutic and regenerative agents to obtain satisfied clinical outcomes [124]. Minimally invasive delivered approaches containing biomaterials, cells, and/or bioactive molecules could complement classic pharmacotherapy, thus finally resulting in pain relief and improving joint function.
Lastly, both a hyaluronic acid (HA) injection and a platelet-rich plasma (PRP) injection may have a remarkable efficacy in the treatment of TMDs. More in detail, as depicted by Harba et al. [30], HA and PRP injections provide greater improvement in patients with TMDs as compared to a HA injection alone.

6. Physical Therapy and Rehabilitation for the Central Sensitization in TMD

6.1. Physical Therapy

Several clinical protocols for interventions and control groups differ; randomized clinical trials (RCTs) of jaw mobilization or stretching conditioning for TMD muscle pain indicate improvements in pain and jaw mobility compared with education and transcranial direct current stimulation, as well as betterment in pain compared with stabilization splints [125,126,127,128,129,130]. Reviews of postural exercises present progress in TMD muscle pain and jaw mobility compared with education alone [28,126,131]. Moreover, combinations of jaw-strengthening and coordination approach, with mobilization and postural programs, enhanced jaw mobility, and reducing joint pain [132]. In this scenario, resistance training with isotonic jaw-opening exercises plays a key role in muscle pain relief and mandibular range of motion improvement [132]. The underlying mechanism appears to be an inhibitory effect on the Golgi tendon. Golgi tendons, located in the target muscle, are stretched by isometric contraction, inducing an inhibitory effect on the muscle activity through Ib muscle fiber [133]. Moreover, postural exercise is typically employed for cervical spine pain management, but it can also be applied in the orofacial region, relieving muscle symptoms such as pain, tension, and stiffness by the influence of the head and mandibular position [134]. It is believed that the incorrect head position can induce muscle pain due to the acceleration of muscle activity in the neck and jaw muscles, as well as postural reflexes [135].

6.2. Occlusal Splints

The difficult relationship between occlusal interferences and temporomandibular disturbance seems to be explained on an animal model, such as the NMDA antagonist MK801 can attenuate occlusal interference-induced hyperalgesia, which suggests that central sensitization mechanisms are involved in the maintenance of the occlusal interference–TMD association [136]. In fact, Xie et al. [136] reported that occlusal interference could directly cause long-term masticatory muscle response in a laboratory animal model. Whether this mechanism may account for cases of TMD in humans needs further investigation. In this scenario, there were no inflammatory cells present, but Substance P expression in masseter muscles of both sides peaked at day 5 and then gradually decreased to the level of the control [137,138]. Their study suggests that, although no evidence of muscle damage and inflammation was found, peripheral sensitization appears to be involved in the mechanism of the EOI-induced masticatory muscle response [139]. However, the peripheral sensitization of nociceptive neurons cannot fully account for the long-standing nociceptive responses of masticatory muscles; a central sensitization mechanism may also be involved [136,140].

6.3. Extracorporeal Shockwave Therapy

The radial ESWT is a pneumatic pressure physical agent modality with direct mechanical stimulation that develops the maximum energy on the skin surface and radially diffuses into the tissues that might be used for musculoskeletal pain relief [34]. Radial ESWT has been widely recognized as a biological modulator that results in the differentiation of mesenchymal stem cells, neovascularization, and release of angiogenetic factors [141]. To date, it is unclear how ESWT can affect temporomandibular disorders. Taking as valid the hypothesis of the mechanotransducive effect of ESWT in other diseases, it could be hypothesized that the waves at the level of the microcirculation can increase the perfusion, promote angiogenesis, and alter the signaling of pain in ischemic tissues caused by the influx of calcium [34,142]. On the other hand, recent articles have shown that free nerve endings degenerate after the application of ESWT and that ESWT produces a transient dysfunction of nerve excitability at the neuromuscular junction, resulting in the downstream of AChR [143]. Although this test was performed on spastic muscles, it could also be extrapolated to the MTP and the energy crisis hypothesis [144]. Lastly, following a purely mechanistic approach, shockwaves may be able to break actin-myosin bonds, as they propagate perpendicular to the sarcomere contractions [143].

6.4. Laser Therapy

Low-level laser therapy (LLLT) has recently been put under the spotlight, because the proponents claim its easy application, limited treatment time, and minimum contraindication. Theoretically, LLLT is a nonthermal type of light, thought to reduce inflammation through the increase of ATP production, improvement of local microcirculation, reduction of edema through an increase of lymphatic flow, and decrease of the prostaglandin E2 and cyclooxygenase-2 levels, albeit the mechanism underlying the therapeutic effects of LLLT is still under debate [145]. Actually, it has a complex mechanism of action, resulting in three main effects on tissues- through direct irradiation without causing a thermal response [146]. Biostimulation occurs through metabolic activation and increased vascularization and fibroblast formation, while the anti-inflammatory and analgesic effects of LLLT are probably due to multiple actions [147,148]. It increases the beta-endorphin level in spinal liquor and increases the urinary excretion of glucocorticoids, which are inhibitors of the synthesis of beta-endorphins [149,150]. It also increases the pressure pain threshold through a complex electrolytic nerve fiber-blocking mechanism and causes a decrease in the release of histamine and acetylcholine and a decrease in the synthesis of bradykinin [151].

6.5. Transcutaneous Electrical Nerve Stimulation

TENS is defined as the application of electrical stimulation to the skin for pain control. It is a well-known form of physical therapy, which is useful for the relief of pain. It is a safe, noninvasive, effective, and swift method of analgesia, and the potential adverse reactions of other methods of pain control are eliminated [152]. Particularly, at the spinal level, low-frequency low-amplitude TENS works on µ receptors, while high-frequency high-amplitude TENS works on δ receptors. Spinal administration in an animal model of a low dose of naloxone (at a low dose, naloxone works as a specific antagonist of the µ receptor of endogenous opioids) and naltrindole (antagonist of δ receptors) in arthritic rats prevented anti-hyperalgesia after both low-frequency low-amplitude and high-frequency high-amplitude TENS showed that the δ and µ receptors were the target of the stimulation [153,154]. Moreover, a particular type of TENS has been used for a long time in dentistry for a variety of purposes, ultralow-frequency TENS (ULFTENS), because of the frequency of the stimulation (0.66 Hz) belonging to the field of ultralow frequencies (<20 Hz) [155]. In the “normal” condition, collaboration exists for the control of arousal between the cortical and subcortical centers [156]. Information transmitted through sensory ULFTENS reaches the nuclear trigeminal sensory complex and, through the latter, is projected to the subcortical areas that control arousal. Acute stress and pain lead to increased arousal (allostasis), followed by the temporary activation of peripheral responses mediated by the autonomous nervous system, as well as the inflammatory, immune, hormonal, and neuromuscular systems [157,158]. It is probable that such action takes place by the “inhibition of the inhibition” of the “activation system”, according to the hypothesis of Thayer [24]. Thus, ULFTENS acts through the balance of the subcortical arousal circuit by enhancing the inhibition through the endorphin system and reducing the cortical activation induced by stress or pain [24].

6.6. Biofeedback

In recent years, the usefulness of biofeedback therapy in patients with different muscle disorders, including TMD, was suggested. Florjanski et al. [28] conducted a systematic review to evaluate the efficacy of biofeedback and concluded that the majority of the included studies presented a significant correlation between biofeedback usage and the reduction of muscle activity.

7. Interventional Therapies and Central Sensitization in TMD

7.1. Acupuncture and Dry Needling

Since the introduction of acupuncture therapy into modern Western medicine, numerous studies have been carried out to investigate and explain the scientific basis behind it [159]. The arrival of qi or “de qi” refers to the transmission of a needling sensation along the meridians, which is often described by the patients as soreness, numbness, fullness, warm sensations, or aching as a result of needle manipulation [160]. Recent histological evidence using rat models seems to suggest this needle grasp sensation is the result of collagen and elastic fibers tightening around the needle during needle manipulation [161]. The authors went further to postulate this mechanical coupling between the needle and soft tissue as being responsible for transducing mechanical signals into fibroblasts and other cells, with resultant therapeutic downstream effects [160,162]. The authors proposed that the stimulation of acupuncture points can relieve pain by causing “hyperstimulation analgesia”, which can be explained by the concept of the “gate control theory of pain”, proposing that the activation of A-δ and C afferent fibers through acupuncture point stimulation sends signals to the spinal cord with a local release of dynorphin and enkephalins [162]. In this scenario, neurotransmitters such as serotonin, dopamine, and norepinephrine are produced, causing the pre- and postsynaptic inhibition of pain transmission, and when the signals reach the hypothalamus and pituitary gland, adrenocorticotropic hormones and endorphins may be produced [163].

7.2. Botulinum Toxin

Botulinum toxin (BoNT) is the protein group produced by anaerobic bacteria called Clostridium botulinum, which has approximately 40 subtypes. However, seven serotypes are typically noted based on antigen specificity. BoNT-A has been the subject of innumerous studies to confirm its antinociceptive effect [164]. Indeed, for a long time, the analgesic effect of BoNT type A (BoNT-A) was considered to be due to the effect of muscle relaxation, particularly in the case of stroke spasticity [165,166]. However, BoNT has been used for neuropathic pain with an analgesic effect independent of muscle relaxation by demonstrating dissociation of the duration of muscle relaxation and duration of pain relief [167,168]. More in detail, the reduction of inflammatory hyper-nociception may be due to an inhibition in the release of certain pain-related neurotransmitters and proinflammatory cytokines [169]. Moreover, the BoNT-A mechanism of action could be not only restricted to a peripheral mechanism but also to a central action on three neurotransmitters: SP, CGRP, and glutamate (Glu), where the inhibition of Glu release takes on a more important role than the one earned peripherally [170]. In vitro models, using cultures of embryonic rat dorsal root ganglion, demonstrated BoNT-A inhibition of SP release and the reduction of stimulated CGRP [171]. Specifically, BoNT-A can directly decrease the amount of CGRP released from trigeminal sensory neurons in cultures of rat trigeminal ganglia. CGRP is a multifunctional regulatory neuropeptide strongly related in the underlying pathology of migraines [172]. In addition, it was found that albumin-induced arthritis increased the release of the proinflammatory cytokines IL1-β and TNF-α [173]. Despite these studies providing important contributions to the better understanding of the antinociceptive mechanism of BoNT-A, more experiments to elucidate this effect are necessary [168].

7.3. Oxygen–Ozone Therapy

Oxygen–ozone therapy is an adjuvant treatment that plays an anti-inflammatory and analgesic effect in several pathological musculoskeletal disorders characterized by chronic inflammatory processes (e.g., low back pain, osteoarthritis, cervical pain, fibromyalgia, and TMD) [36]. The effect of oxygen–ozone therapy mimics an acute oxidative stress that, if properly balanced, is not harmful but is able to provoke positive biological responses and reverse chronic oxidative stress (degenerative process, aging, etc.) [174]. This hypothesis about ozone and oxidative stress modulation could be better defined as a “real non-toxic therapeutic shock able to restore homeostasis” [175]. Low doses of oxygen–ozone could therefore play a role in the regulation of prostaglandin synthesis, in the release of bradykinin, and in the increase of macrophage and leukocyte secretions. It is widely accepted that pain is a common symptom related to the inflammatory process, and oxygen–ozone therapy could play a key role not only in the management of inflammation but also in nociceptive perception and modulation [176]. As for the analgesic use, after the administration of oxygen–ozone, an increase in the antioxidant molecules (serotonin and endogenous opioids) has been demonstrated, which would induce pain relief by stimulating the antinociceptive pathways [177,178].

8. Study Limitations and Strengths

In conclusion, the diagnosis was a fundamental starting point to comprehend the pathology of these subjects; indeed, although all of them presented craniofacial pain, it varied depending on the origin: muscular, arthrogenous, or a combination of them. Chronic craniofacial muscle pain associated with TMD involves multiple peripheral and central mechanisms, and it should be taken into consideration that the coronavirus pandemic has caused significant adverse effects on their psycho-emotional status, resulting in the intensification of their bruxism and TMD symptoms and thus leading to increased orofacial pain [179,180].
This study is not free from limitations, such as the lack of systematic research of the literature and the absence of a meta-analysis. However, the study heterogeneity might not allow a quantitative analysis, in accordance with the Cochrane Handbook for Systematic Review of Intervention (Ver, 6.2, 2021) [181]. Furthermore, it could be difficult to draw strong conclusions starting from so wide a presence of observational studies with different outcomes measured, testifying to the difficulty in the assessment of the central sensitization.
On the other hand, it should be noted that this comprehensive review is the first in the scientific literature investigating both the diagnosis and treatment of patients with TMD by adequate control of the central sensitization. The resulting evidence showed that several pharmacological and conservative approaches could have a potential effective role in the regulation of the central sensitization in patients affected by TMD pain.

9. Conclusions

Taken together, the findings of the present comprehensive review showed that the central sensitization and the inhibitory system of descending pain might play a role in the TMD clinical pattern. In this context, pharmacological drugs and conservative approaches (e.g., occlusal splints, ESWT, LLLT, TENS, and oxygen–ozone therapy) could have a positive impact in terms of the central sensitization of TMD pain. Further observational studies should investigate the role of these rehabilitative approaches in pain relief in patients affected by TMD.

Author Contributions

Conceptualization, M.F., P.F. and A.d.S.; methodology, A.G., A.A. and A.d.S.; validation, M.F., N.M. and D.D.V.; investigation, M.F., N.M. and F.F.; data curation, N.M., F.F. and A.d.S.; writing—original draft preparation, M.F., N.M. and F.F.; writing—review and editing, A.G. and A.d.S.; visualization, D.D.V., A.A. and P.F; and supervision, A.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Dario Calafiore for his support of this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schiffman, E.; Ohrbach, R.; Truelove, E.; Look, J.; Anderson, G.; Goulet, J.P.; List, T.; Svensson, P.; Gonzalez, Y.; Lobbezoo, F.; et al. Diagnostic Criteria for Temporomandibular Disorders (DC/TMD) for Clinical and Research Applications: Recommendations of the International RDC/TMD Consortium Network* and Orofacial Pain Special Interest Group†. J. Oral Facial Pain Headache 2014, 28, 6–27. [Google Scholar] [CrossRef]
  2. Bueno, C.H.; Pereira, D.D.; Pattussi, M.P.; Grossi, P.K.; Grossi, M.L. Gender differences in temporomandibular disorders in adult populational studies: A systematic review and meta-analysis. J. Oral Rehabil. 2018, 45, 720–729. [Google Scholar] [CrossRef]
  3. Simoen, L.; Van den Berghe, L.; Jacquet, W.; Marks, L. Depression and anxiety levels in patients with temporomandibular disorders: Comparison with the general population. Clin. Oral Investig. 2020, 24, 3939–3945. [Google Scholar] [CrossRef]
  4. Bitiniene, D.; Zamaliauskiene, R.; Kubilius, R.; Leketas, M.; Gailius, T.; Smirnovaite, K. Quality of life in patients with temporomandibular disorders. A systematic review. Stomatologija 2018, 20, 3–9. [Google Scholar]
  5. Castro-Calderón, A.; Roccuzzo, A.; Ferrillo, M.; Gada, S.; González-Serrano, J.; Fonseca, M.; Molinero-Mourelle, P. Hyaluronic acid injection to restore the lost interproximal papilla: A systematic review. Acta Odontol Scand. 2022, 80, 295–307. [Google Scholar] [CrossRef]
  6. Baad-Hansen, L.; Benoliel, R. Neuropathic orofacial pain: Facts and fiction. Cephalalgia 2017, 37, 670–679. [Google Scholar] [CrossRef] [Green Version]
  7. Nicholas, M.; Vlaeyen, J.W.S.; Rief, W.; Barke, A.; Aziz, Q.; Benoliel, R.; Cohen, M.; Evers, S.; Giamberardino, M.A.; Goebel, A.; et al. The IASP classification of chronic pain for ICD-11: Chronic primary pain. Pain 2019, 160, 28–37. [Google Scholar] [CrossRef]
  8. Treede, R.D.; Rief, W.; Barke, A.; Aziz, Q.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Evers, S.; Finnerup, N.B.; First, M.B.; et al. Chronic pain as a symptom or a disease: The IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain 2019, 160, 19–27. [Google Scholar] [CrossRef] [Green Version]
  9. Moreno-Fernández, A.M.; Jiménez-Castellanos, E.; Iglesias-Linares, A.; Bueso-Madrid, D.; Fernández-Rodríguez, A.; de Miguel, M. Fibromyalgia syndrome and temporomandibular disorders with muscular pain. A review. Mod. Rheumatol. 2017, 27, 210–216. [Google Scholar] [CrossRef]
  10. Ferrillo, M.; Migliario, M.; Marotta, N.; Fortunato, F.; Bindi, M.; Pezzotti, F.; Ammendolia, A.; Giudice, A.; Foglio Bonda, P.L.; de Sire, A. Temporomandibular disorders and neck pain in primary headache patients: A retrospective machine learning study. Acta Odontol. Scand. 2022, 29, 1–7. [Google Scholar] [CrossRef]
  11. Chaves, T.C.; Dach, F.; Florencio, L.L.; Carvalho, G.F.; Gonçalves, M.C.; Bigal, M.E.; Speciali, J.G.; Bevilaqua-Grossi, D. Concomitant Migraine and Temporomandibular Disorders are Associated With Higher Heat Pain Hyperalgesia and Cephalic Cutaneous Allodynia. Clin. J. Pain 2016, 32, 882–888. [Google Scholar] [CrossRef]
  12. Greenspan, J.D.; Slade, G.D.; Bair, E.; Dubner, R.; Fillingim, R.B.; Ohrbach, R.; Knott, C.; Diatchenko, L.; Liu, Q.; Maixner, W. Pain sensitivity and autonomic factors associated with development of TMD: The OPPERA prospective cohort study. J. Pain 2013, 14, T63–T74.e746. [Google Scholar] [CrossRef] [Green Version]
  13. Paolucci, T.; de Sire, A.; Ferrillo, M.; di Fabio, D.; Molluso, A.; Patruno, A.; Pesce, M.; Lai, C.; Ciacchella, C.; Saggino, A.; et al. Telerehabilitation proposal of mind-body technique for physical and psychological outcomes in patients with fibromyalgia. Front. Physiol. 2022, 13, 917956. [Google Scholar] [CrossRef]
  14. Woolf, C.J. Central sensitization: Implications for the diagnosis and treatment of pain. Pain 2011, 152, S2–S15. [Google Scholar] [CrossRef]
  15. Adams, L.M.; Turk, D.C. Central sensitization and the biopsychosocial approach to understanding pain. J. Appl. Behav. Res. 2018, 23, e12125. [Google Scholar] [CrossRef]
  16. Merrill, R.L. Central mechanisms of orofacial pain. Dent. Clin. N. Am. 2007, 51, 45–59. [Google Scholar] [CrossRef]
  17. Craig, A.D. A new view of pain as a homeostatic emotion. Trends Neurosci. 2003, 26, 303–307. [Google Scholar] [CrossRef]
  18. Sessle, B.J. Peripheral and central mechanisms of orofacial pain and their clinical correlates. Minerva Anestesiol. 2005, 71, 117–136. [Google Scholar]
  19. Merskey, H.; Bogduk, N. Classification of Chronic Pain, 2nd ed.; International Association for the Study of Pain Press: Seattle, WA, USA, 1994; p. 210. [Google Scholar]
  20. Lorduy, K.M.; Liegey-Dougall, A.; Haggard, R.; Sanders, C.N.; Gatchel, R.J. The prevalence of comorbid symptoms of central sensitization syndrome among three different groups of temporomandibular disorder patients. Pain Pract. 2013, 13, 604–613. [Google Scholar] [CrossRef]
  21. Dahan, H.; Shir, Y.; Velly, A.; Allison, P. Specific and number of comorbidities are associated with increased levels of temporomandibular pain intensity and duration. J. Headache Pain 2015, 16, 528. [Google Scholar] [CrossRef] [Green Version]
  22. Sanzarello, I.; Merlini, L.; Rosa, M.A.; Perrone, M.; Frugiuele, J.; Borghi, R.; Faldini, C. Central sensitization in chronic low back pain: A narrative review. J. Back Musculoskelet Rehabil. 2016, 29, 625–633. [Google Scholar] [CrossRef] [PubMed]
  23. Monaco, A.; Cattaneo, R.; Marci, M.C.; Pietropaoli, D.; Ortu, E. Central Sensitization-Based Classification for Temporomandibular Disorders: A Pathogenetic Hypothesis. Pain Res. Manag. 2017, 2017, 5957076. [Google Scholar] [CrossRef] [PubMed]
  24. de Souza, R.F.; Lovato da Silva, C.H.; Nasser, M.; Fedorowicz, Z.; Al-Muharraqi, M.A. Interventions for the management of temporomandibular joint osteoarthritis. Cochrane Database Syst. Rev. 2012, 2012, CD007261. [Google Scholar]
  25. Ferrillo, M.; Ammendolia, A.; Paduano, S.; Calafiore, D.; Marotta, N.; Migliario, M.; Fortunato, L.; Giudice, A.; Michelotti, A.; de Sire, A. Efficacy of rehabilitation on reducing pain in muscle-related temporomandibular disorders: A systematic review and meta-analysis of randomized controlled trials. J. Back Musculoskelet Rehabil. 2022, 18, 1–16. [Google Scholar] [CrossRef]
  26. Ferrillo, M.; Nucci, L.; Giudice, A.; Calafiore, D.; Marotta, N.; Minervini, G.; d’Apuzzo, F.; Ammendolia, A.; Perillo, L.; de Sire, A. Efficacy of conservative approaches on pain relief in patients with temporomandibular joint disorders: A systematic review with network meta-analysis. Cranio 2022, 23, 1–17. [Google Scholar] [CrossRef]
  27. Armijo-Olivo, S.; Pitance, L.; Singh, V.; Neto, F.; Thie, N.; Michelotti, A. Effectiveness of Manual Therapy and Therapeutic Exercise for Temporomandibular Disorders: Systematic Review and Meta-Analysis. Phys. Ther. 2016, 96, 9–25. [Google Scholar] [CrossRef] [Green Version]
  28. Florjanski, W.; Malysa, A.; Orzeszek, S.; Smardz, J.; Olchowy, A.; Paradowska-Stolarz, A.; Wieckiewicz, M. Evaluation of Biofeedback Usefulness in Masticatory Muscle Activity Management—A Systematic Review. J. Clin. Med. 2019, 8, 766. [Google Scholar] [CrossRef] [Green Version]
  29. Andre, A.; Kang, J.; Dym, H. Pharmacologic Treatment for Temporomandibular and Temporomandibular Joint Disorders. Oral Maxillofac. Surg. Clin. N. Am. 2022, 34, 49–59. [Google Scholar] [CrossRef]
  30. Harba, A.N.; Harfoush, M. Evaluation of the participation of hyaluronic acid with platelet-rich plasma in the treatment of temporomandibular joint disorders. Dent. Med. Probl. 2021, 58, 81–88. [Google Scholar] [CrossRef]
  31. Deregibus, A.; Ferrillo, M.; Grazia Piancino, M.; Chiara Domini, M.; de Sire, A.; Castroflorio, T. Are occlusal splints effective in reducing myofascial pain in patients with muscle-related temporomandibular disorders? A randomized-controlled trial. Turk. J. Phys. Med. Rehabil. 2021, 67, 32–40. [Google Scholar] [CrossRef]
  32. Kuzmanovic Pficer, J.; Dodic, S.; Lazic, V.; Trajkovic, G.; Milic, N.; Milicic, B. Occlusal stabilization splint for patients with temporomandibular disorders: Meta-analysis of short and long term effects. PLoS ONE 2017, 12, e0171296. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, G.Z.; Jia, J.; Jin, L.; Li, J.H.; Wang, Z.Y.; Cao, D.Y. Low-Level Laser Therapy for Temporomandibular Disorders: A Systematic Review with Meta-Analysis. Pain Res. Manag. 2018, 2018, 4230583. [Google Scholar] [CrossRef] [Green Version]
  34. Marotta, N.; Ferrillo, M.; Demeco, A.; Drago Ferrante, V.; Inzitari, M.T.; Pellegrino, R.; Pino, I.; Russo, I.; de Sire, A.; Ammendolia, A. Effects of Radial Extracorporeal Shock Wave Therapy in Reducing Pain in Patients with Temporomandibular Disorders: A Pilot Randomized Controlled Trial. Appl. Sci. 2022, 12, 3821. [Google Scholar] [CrossRef]
  35. Fertout, A.; Manière-Ezvan, A.; Lupi, L.; Ehrmann, E. Management of temporomandibular disorders with transcutaneous electrical nerve stimulation: A systematic review. Cranio 2022, 40, 217–228. [Google Scholar] [CrossRef]
  36. de Sire, A.; Marotta, N.; Ferrillo, M.; Agostini, F.; Sconza, C.; Lippi, L.; Respizzi, S.; Giudice, A.; Invernizzi, M.; Ammendolia, A. Oxygen-Ozone Therapy for Reducing Pro-Inflammatory Cytokines Serum Levels in Musculoskeletal and Temporomandibular Disorders: A Comprehensive Review. Int. J. Mol. Sci. 2022, 23, 2528. [Google Scholar] [CrossRef] [PubMed]
  37. Yin, Y.; He, S.; Xu, J.; You, W.; Li, Q.; Long, J.; Luo, L.; Kemp, G.J.; Sweeney, J.A.; Li, F.; et al. The neuro-pathophysiology of temporomandibular disorders-related pain: A systematic review of structural and functional MRI studies. J. Headache Pain 2020, 19, 21–78. [Google Scholar] [CrossRef]
  38. Bender, S.D. Orofacial pain and headache: A review and look at the commonalities. Curr Pain Headache Rep. 2014, 18, 400. [Google Scholar] [CrossRef] [PubMed]
  39. Latremoliere, A.; Woolf, C.J. Central sensitization: A generator of pain hypersensitivity by central neural plasticity. J. Pain 2009, 10, 895–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Cairns, B.E.; Sessle, B.J.; Hu, J.W. Activation of peripheral GABAA receptors inhibits temporomandibular joint-evoked jaw muscle activity. J. Neurophysiol. 1999, 81, 1966–1969. [Google Scholar] [CrossRef] [Green Version]
  41. Tashiro, A.; Bereiter, D.A.; Thompson, R.; Nishida, Y. GABAergic influence on temporomandibular joint-responsive spinomedullary neurons depends on estrogen status. Neuroscience 2014, 259, 53–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sperry, M.M.; Granquist, E.J.; Winkelstein, B.A. Increased substance P and synaptic remodeling occur in the trigeminal sensory system with sustained osteoarthritic temporomandibular joint sensitivity. Pain Rep. 2021, 6, e911. [Google Scholar] [CrossRef] [PubMed]
  43. Costa, Y.M.; Conti, P.C.; de Faria, F.A.; Bonjardim, L.R. Temporomandibular disorders and painful comorbidities: Clinical association and underlying mechanisms. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2017, 123, 288–297. [Google Scholar] [CrossRef] [PubMed]
  44. Conti, P.C.; Costa, Y.M.; Gonçalves, D.A.; Svensson, P. Headaches and myofascial temporomandibular disorders: Overlapping entities, separate managements? J. Oral Rehabil. 2016, 43, 702–715. [Google Scholar] [CrossRef]
  45. Campi, L.B.; Visscher, C.M.; Ongaro, P.C.J.; do Vale Braido, G.V.; Fernandes, G.; Gonçalves, D.A.G. Widespread Pain and Central Sensitization in Adolescents with Signs of Painful Temporomandibular Disorders. J. Oral Facial. Pain Headache 2020, 34, 83–91. [Google Scholar] [CrossRef] [PubMed]
  46. Lim, P.F.; Smith, S.; Bhalang, K.; Slade, G.D.; Maixner, W. Development of temporomandibular disorders is associated with greater bodily pain experience. Clin. J. Pain 2010, 26, 116–120. [Google Scholar] [CrossRef] [Green Version]
  47. Dalewski, B.; Kamińska, A.; Kiczmer, P.; Węgrzyn, K.; Pałka, Ł.; Janda, K.; Sobolewska, E. Pressure Algometry Evaluation of Two Occlusal Splint Designs in Bruxism Management-Randomized, Controlled Clinical Trial. J. Clin. Med. 2021, 10, 2342. [Google Scholar] [CrossRef]
  48. Gomes, M.B.; Guimarães, J.P.; Guimarães, F.C.; Neves, A.C. Palpationand pressure pain threshold: Reliability and validity in patientswith temporomandibular disorders. Cranio 2008, 26, 202–210. [Google Scholar] [CrossRef]
  49. Fernández-de-las-Peñas, C.; Galán-del-Río, F.; Ortega-Santiago, R.; Jiménez-García, R.; Arendt-Nielsen, L.; Svensson, P. Bilateral thermal hyperalgesia in trigeminal and extra-trigeminal regions in patients with myofascial temporomandibular disorders. Exp. Brain Res. 2010, 202, 171–179. [Google Scholar] [CrossRef]
  50. Chesterton, L.S.; Sim, J.; Wright, C.C.; Foster, N.E. Interrater reliability of algometry in measuring pressure pain thresholds in healthy humans; using multiple raters. Clin. J. Pain 2007, 23, 760–766. [Google Scholar] [CrossRef]
  51. Bernhardt, O.; Schiffman, E.L.; Look, J.O. Reliability and validity of a new fingertip-shaped pressure algometer for assessing pressure pain thresholds in the temporomandibular joint and masticatory muscles. J. Orofac. Pain. 2007, 21, 29–38. [Google Scholar]
  52. Treede, R.D.; Rolke, R.; Andrews, K.; Magerl, W. Pain elicited byblunt pressure: Neurobiological basis and clinical relevance. Pain 2002, 98, 235–240. [Google Scholar] [CrossRef]
  53. List, T.; Helkimo, M.; Karlsson, R. Influence of pressure rates onthe reliability of a pressure threshold meter. J. Craniomandib. Disord. 1991, 5, 173–178. [Google Scholar] [PubMed]
  54. Farella, M.; Michelotti, A.; Cimino, R.; Martina, R. An investigation of central and peripheral factors affecting pressure pain thresholds of the human jaw muscles. J. Musculoskel. Pain 1999, 7, 253–259. [Google Scholar] [CrossRef]
  55. da Silva, M.M.; Albertini, R.; Leal-Junior, E.C.; de Tarso Camillo de Carvalho, P.; Silva, J.A., Jr.; Bussadori, S.K.; de Oliveira, L.V.; Casarin, C.A.; Andrade, E.L.; Bocalini, D.S.; et al. Effects of exercise training and photobiomodulation therapy (EXTRAPHOTO) on pain in women with fibromyalgia and temporomandibular disorder: Study protocol for a randomized controlled trial. Trials 2015, 16, 252. [Google Scholar] [CrossRef]
  56. Herpich, C.M.; Leal-Junior, E.C.P.; Amaral, A.P.; Tosato, J.; Glória, I.P.; Garcia, M.B.; Barbosa, B.R.; El Hage, Y.; Arruda, É.E.; Gomes, C.Á.; et al. Effects of phototherapy on muscle activity and pain in individuals with temporomandibular disorder: A study protocol for a randomized controlled trial. Trials 2014, 15, 491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Svensson, P.; Baad-Hansen, L.; Pigg, M.; List, T.; Eliav, E.; Ettlin, D.; Michelotti, A.; Tsukiyama, Y.; Matsuka, Y.; Jääskeläinen, S.K.; et al. Guidelines and recommendations for assessment of somatosensory function in oro-facial pain conditions—A taskforce report. J. Oral Rehabil. 2011, 38, 366–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Hansson, P.; Backonja, M.; Bouhassira, D. Usefulness andlimitations of quantitative sensory testing: Clinical andresearch application in neuropathic pain states. Pain 2007, 129, 256–259. [Google Scholar] [CrossRef]
  59. Cruccu, G.; Sommer, C.; Anand, P.; Attal, N.; Baron, R.; Garcia-Larrea, L.; Haanpaa, M.; Jensen, T.S.; Serra, J.; Treede, R.D. EFNS guidelines on neuropathic pain assessment: Revised 2009. Eur. J. Neurol. 2010, 17, 1010–1018. [Google Scholar] [CrossRef]
  60. Maier, C.; Baron, R.; Tolle, T.R.; Binder, A.; Birbaumer, N.; Birklein, F.; Gierthmühlen, J.; Flor, H.; Geber, C.; Huge, V. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): Somatosensory abnormalities in 1236 patients with different neuropathicpain syndromes. Pain 2010, 150, 439–450. [Google Scholar] [CrossRef]
  61. Akinci, A.; Al Shaker, M.; Chang, M.H.; Cheung, C.W.; Danilov, A.; José Dueñas, H.; Kim, Y.C.; Guillen, R.; Tassanawipas, W.; Treuer, T.; et al. Predictive factors and clinical biomarkers for treatment in patients with chronic pain caused by osteoarthritis with a central sensitisation component. Int. J. Clin. Pract. 2016, 70, 31–44. [Google Scholar] [CrossRef] [Green Version]
  62. Pigg, M.; Baad-Hansen, L.; Svensson, P.; Drangsholt, M.; List, T. Reliability of intraoral quantitative sensory testing (QST). Pain 2010, 148, 220–226. [Google Scholar] [CrossRef] [Green Version]
  63. Plesh, O.; Adams, S.H.; Gansky, S.A. Temporomandibular joint and muscle disorder-type pain and comorbid pains in a national US sample. J. Orofac. Pain 2011, 25, 190–198. [Google Scholar]
  64. Botros, J.; Gornitsky, M.; Samim, F.; der Khatchadourian, Z.; Velly, A.M. Back and neck pain: A comparison between acute and chronic pain-related Temporomandibular Disorders. Can. J. Pain 2022, 6, 112–120. [Google Scholar] [CrossRef]
  65. Ohrbach, R.; Fillingim, R.B.; Mulkey, F.; Gonzalez, Y.; Gordon, S.; Gremillion, H.; Lim, P.-F.; Ribeiro-Dasilva, M.; Greenspan, J.D.; Knott, C. Clinical findings and pain symptoms as potential risk factors for chronic tmd: Descriptive data and empirically identified domains from the opera case-control study. J. Pain 2011, 12, T27–T45. [Google Scholar] [CrossRef] [Green Version]
  66. Headache Classification Committee of the International Headache Society (IHS). The International Classification of Headache Disorders; 3rd edition (beta version). Cephalalgia 2013, 33, 629–808. [Google Scholar] [CrossRef] [PubMed]
  67. Matre, D.; Knardahl, S. ‘Central sensitization’ in chronic neck/shoulder pain. Scand. J. Pain 2012, 3, 230–235. [Google Scholar] [CrossRef] [PubMed]
  68. Su, M.; Yu, S. Chronic migraine: A process of dysmodulation and sensitization. Mol. Pain 2018, 14, 1744806918767697. [Google Scholar] [CrossRef]
  69. Gonçalves, D.A.; Camparis, C.M.; Speciali, J.G.; Franco, A.L.; Castanharo, S.M.; Bigal, M.E. Temporomandibular disorders are differentially associated with headache diagnoses: A controlled study. Clin. J. Pain 2011, 27, 611–615. [Google Scholar] [CrossRef]
  70. Furquim, B.D.; Flamengui, L.M.; Conti, P.C. TMD and chronic pain: A current view. Dental Press J. Orthod. 2015, 20, 127–133. [Google Scholar] [CrossRef]
  71. Bevilaqua-Grossi, D.; Lipton, R.B.; Napchan, U.; Grosberg, B.; Ashina, S.; Bigal, M.E. Temporomandibular disorders and cutaneous allodynia are associated in individuals with migraine. Cephalalgia 2010, 30, 425–432. [Google Scholar] [CrossRef]
  72. Mayer, T.G.; Neblett, R.; Cohen, H.; Howard, K.J.; Choi, Y.H.; Williams, M.J.; Perez, Y.; Gatchel, R.J. The development and psychometric validation of the central sensitization inventory. Pain Pract. 2012, 12, 276–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Nijs, J.; Torres-Cueco, R.; van Wilgen, C.P.; Girbes, E.L.; Struyf, F.; Roussel, N.; van Oosterwijck, J.; Daenen, L.; Kuppens, K.; Vanwerweeen, L.; et al. Applying modern pain neuroscience in clinical practice: Criteria for the classification of central sensitization pain. Pain Physician 2014, 17, 447–457. [Google Scholar] [CrossRef] [PubMed]
  74. Vandyken, B.; Keizer, A.; Vandyken, C.; Macedo, L.G.; Kuspinar, A.; Dufour, S. Pelvic floor muscle tenderness on digital palpation among women: Convergent validity with central sensitization. Braz. J. Phys. Ther. 2020, 25, 256–261. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, S.H.; Yoon, K.B.; Yoon, D.M.; Yoo, J.H.; Ahn, K.R. Influence of centrally mediated symptoms on postoperative pain in osteoarthritis patients undergoing total knee arthroplasty: A prospective observational evaluation. Pain Pract. 2015, 15, E46–E53. [Google Scholar] [CrossRef]
  76. Neblett, R.; Hartzell, M.M.; Williams, M.; Bevers, K.R.; Mayer, T.G.; Gatchel, R.J. Use of the Central Sensitization Inventory (CSI) as a treatment outcome measure for patients with chronic spinal pain disorder in a functional restoration program. Spine J. 2017, 17, 819–829. [Google Scholar] [CrossRef]
  77. Beales, D.; Fary, R.; Little, C.; Nambiar, S.; Sveinall, H.; Yee, Y.L.; Tampin, B.; Mitchell, T. Characterisation of pain in people with hereditary neuropathy with liability to pressure palsy. J. Neurol. 2017, 264, 2464–2471. [Google Scholar] [CrossRef] [Green Version]
  78. Ruscheweyh, R.; Marziniak, M.; Stumpenhorst, F.; Reinholz, J.; Knecht, S. Pain sensitivity can be assessed by self-rating: Development and validation of the Pain Sensitivity Questionnaire. Pain 2009, 146, 65–74. [Google Scholar] [CrossRef]
  79. Coronado, R.A.; George, S.Z. The Central Sensitization Inventory and Pain Sensitivity Questionnaire: An exploration of construct validity and associations with widespread pain sensitivity among individuals with shoulder pain. Musculoskelet Sci. Pract. 2018, 36, 61–67. [Google Scholar] [CrossRef] [PubMed]
  80. Ruscheweyh, R.; Verneuer, B.; Dany, K.; Marziniak, M.; Wolowski, A.; Colak-Ekici, R.; Schulte, T.L.; Bullmann, V.; Grewe, S.; Gralow, I.; et al. Validation of the pain sensitivity questionnaire in chronic pain patients. Pain 2012, 153, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  81. den Boer, C.; Dries, L.; Terluin, B.; van der Wouden, J.C.; Blankenstein, A.H.; van Wilgen, C.P.; Lucassen, P.; van der Horst, H.E. Central sensitization in chronic pain and medically unexplained symptom research: A systematic review of definitions; operationalizations and measurement instruments. J. Psychosom. Res. 2019, 117, 32–40. [Google Scholar] [CrossRef]
  82. Dixon, E.A.; Benham, G.; Sturgeon, J.A.; Mackey, S.; Johnson, K.A.; Younger, J. Development of the Sensory Hypersensitivity Scale (SHS): A self-report tool for assessing sensitivity to sensory stimuli. J. Behav. Med. 2016, 39, 537–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. La Touche, R.; Paris-Alemany, A.; Hidalgo-Pérez, A.; López-de-Uralde-Villanueva, I.; Angulo-Diaz-Parreño, S.; Muñoz-García, D. Evidence for Central Sensitization in Patients with Temporomandibular Disorders: A Systematic Review and Meta-analysis of Observational Studies. Pain Pract. 2018, 18, 388–409. [Google Scholar] [CrossRef] [PubMed]
  84. Meng, H.; Dai, J.; Li, Y. Quantitative sensory testing in patients with the muscle pain subtype of temporomandibular disorder: A systemic review and meta-analysis. Clin. Oral Investig. 2021, 25, 6547–6559. [Google Scholar] [CrossRef]
  85. Fernández-de-las-Peñas, C.; Galán-del-Río, F.; Fernández-Carnero, J.; Pesquera, J.; Arendt-Nielsen, L.; Svensson, P. Bilateral widespread mechanical pain sensitivity in women with myofascial temporomandibular disorder: Evidence of impairment in central nociceptive processing. J. Pain 2009, 10, 1170–1178. [Google Scholar] [CrossRef]
  86. Daemen, M.; Kurvers, H.; Kitslaar, P.; Slaaf, D.W.; Bullens, P.H.; Van den Wildenberg, F.A. Neurogenic inflammation in an animal model of neuropathic pain. Neurol. Res. 1998, 20, 41–45. [Google Scholar] [CrossRef]
  87. Svensson, P.; List, T.; Hector, G. Analysis of stimulus-evoked pain in patients with myofascial temporomandibular pain disorders. Pain 2001, 92, 399–409. [Google Scholar] [CrossRef]
  88. Sarlani, E.; Grace, E.G.; Reynolds, M.A.; Greenspan, J.D. Evidence for up-regulated central nociceptive processing in patients with masticatory myofascial pain. J. Orofac. Pain 2004, 18, 41–55. [Google Scholar] [PubMed]
  89. Fernández-de-Las-Peñas, C.; Von Piekartz, H. Clinical Reasoning for the Examination and Physical Therapy Treatment of Temporomandibular Disorders (TMD): A Narrative Literature Review. J. Clin. Med. 2020, 9, 3686. [Google Scholar] [CrossRef]
  90. Campi, L.B.; Jordani, P.C.; Tenan, H.L.; Camparis, C.M.; Gonçalves, D.A. Painful temporomandibular disorders and central sensitization: Implications for management-a pilot study. Int. J. Oral Maxillofac. Surg. 2017, 46, 104–110. [Google Scholar] [CrossRef] [Green Version]
  91. Asquini, G.; Bianchi, A.E.; Borromeo, G.; Locatelli, M.; Falla, D. The impact of COVID-19-related distress on general health, oral behaviour, psychosocial features, disability and pain intensity in a cohort of Italian patients with temporomandibular disorders. PLoS ONE 2021, 16, e0245999. [Google Scholar] [CrossRef]
  92. Ghurye, S.; McMillan, R. Pain-Related Temporomandibular Disorder-Current Perspectives and Evidence-Based Management. Dent. Update 2015, 42, 533–546. [Google Scholar] [CrossRef] [Green Version]
  93. Dworkin, S.F.; Huggins, K.H.; LeResche, L.; Von Korff, M.; Howard, J.; Truelove, E.; Sommers, E. Epidemiology of signs and symptoms in temporomandibular disorders: Clinical signs in cases and controls. J. Am. Dent. Assoc. 1990, 120, 273–281. [Google Scholar] [CrossRef]
  94. Chichorro, J.G.; Porreca, F.; Sessle, B. Mechanisms of craniofacial pain. Cephalalgia 2017, 37, 613–626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Campbell, J.N.; Meyer, R.A. Mechanisms of neuropathic pain. Neuron 2006, 52, 77–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ossipov, M.H.; Dussor, G.O.; Porreca, F. Central modulation of pain. J. Clin. Investig. 2010, 120, 3779–3787. [Google Scholar] [CrossRef] [PubMed]
  97. Takemura, M.; Sugiyo, S.; Moritani, M.; Kobayashi, M.; Yonehara, N. Mechanisms of orofacial pain control in the central nervous system. Arch. Histol. Cytol. 2006, 69, 79–100. [Google Scholar] [CrossRef] [Green Version]
  98. Mujakperuo, H.R.; Watson, M.; Morrison, R.; Macfarlane, T.V. Pharmacological interventions for pain in patients with temporomandibular disorders. Cochrane Database Syst. Rev. 2010, Cd004715. [Google Scholar] [CrossRef]
  99. Haas, D.A. Pharmacologic considerations in the management of temporomandibular disorders. J. Can. Dent. Assoc. 1995, 61, 105–114. [Google Scholar] [PubMed]
  100. Wright, E.F. Manual of Temporomandibular Disorders, 2nd ed.; Wiley-Blackwell: Ames, IA, USA, 2010. [Google Scholar]
  101. Ta, L.E.; Dionne, R.A. Treatment of painful temporomandibular joints with a cyclooxygenase-2 inhibitor: A randomized placebo-controlled comparison of celecoxib to naproxen. Pain 2004, 111, 13–21. [Google Scholar] [CrossRef] [PubMed]
  102. de Carli, M.L.; Guerra, M.B.; Nunes, T.B.; di Matteo, R.C.; de Luca, C.E.; Aranha, A.C.; Bolzan, M.C.; Witzel, A.L. Piroxicam and laser phototherapy in the treatment of TMJ arthralgia: A double-blind randomised controlled trial. J. Oral Rehabil. 2013, 40, 171–178. [Google Scholar] [CrossRef]
  103. Di Rienzo Businco, L.; Di Rienzo Businco, A.; D’Emilia, M.; Lauriello, M.; Coen Tirelli, G. Topical versus systemic diclofenac in the treatment of temporo-mandibular joint dysfunction symptoms. Acta Otorhinolaryngol. Ital. 2004, 24, 279–283. [Google Scholar] [PubMed]
  104. Dong, X.D.; Svensson, P.; Cairns, B.E. The analgesic action of topical diclofenac may be mediated through peripheral NMDA receptor antagonism. Pain 2009, 147, 36–45. [Google Scholar] [CrossRef] [PubMed]
  105. Kulkarni, S.; Thambar, S.; Arora, H. Evaluating the effectiveness of nonsteroidal anti-inflammatory drug(s) for relief of pain associated with temporomandibular joint disorders: A systematic review. Clin. Exp. Dent. Res. 2020, 6, 134–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Jackson, J.L.; Kuriyama, A.; Kuwatsuka, Y.; Nickoloff, S.; Storch, D.; Jackson, W.; Zhang, Z.J.; Hayashino, Y. Beta-blockers for the prevention of headache in adults; a systematic review and meta-analysis. PLoS ONE 2019, 14, e0212785. [Google Scholar] [CrossRef] [PubMed]
  107. Rodrigues, L.L.; Oliveira, M.C.; Pelegrini-da-Silva, A.; de Arruda Veiga, M.C.; Parada, C.A.; Tambeli, C.H. Peripheral sympathetic component of the temporomandibular joint inflammatory pain in rats. J. Pain 2006, 7, 929–936. [Google Scholar] [CrossRef]
  108. Oliveira-Fusaro, M.C.; Clemente-Napimoga, J.T.; Teixeira, J.M.; Torres-Chávez, K.E.; Parada, C.A.; Tambeli, C.H. 5-HT induces temporomandibular joint nociception in rats through the local release of inflammatory mediators and activation of local β adrenoceptors. Pharmacol. Biochem. Behav. 2012, 102, 458–464. [Google Scholar] [CrossRef] [Green Version]
  109. Tchivileva, I.E.; Hadgraft, H.; Lim, P.F.; Di Giosia, M.; Ribeiro-Dasilva, M.; Campbell, J.H.; Willis, J.; James, R.; Herman-Giddens, M.; Fillingim, R.B.; et al. Efficacy and safety of propranolol for treatment of temporomandibular disorder pain: A randomized; placebo-controlled clinical trial. Pain 2020, 161, 1755–1767. [Google Scholar] [CrossRef]
  110. Burch, R. Antidepressants for Preventive Treatment of Migraine. Curr. Treat.Options Neurol. 2019, 21, 18. [Google Scholar] [CrossRef]
  111. Haviv, Y.; Rettman, A.; Aframian, D.; Sharav, Y.; Benoliel, R. Myofascial pain: An open study on the pharmacotherapeutic response to stepped treatment with tricyclic antidepressants and gabapentin. J. Oral Facial. Pain Headache 2015, 29, 144–151. [Google Scholar] [CrossRef]
  112. Finnerup, N.B.; Sindrup, S.H.; Jensen, T.S. The evidence for pharmacological treatment of neuropathic pain. Pain 2010, 150, 573–581. [Google Scholar]
  113. Rizzatti-Barbosa, C.M.; Nogueira, M.T.; de Andrade, E.D.; Ambrosano, G.M.; de Barbosa, J.R. Clinical evaluation of amitriptyline for the control of chronic pain caused by temporomandibular joint disorders. Cranio 2003, 21, 221–225. [Google Scholar] [CrossRef]
  114. Cascos-Romero, J.; Vázquez-Delgado, E.; Vázquez-Rodríguez, E.; Gay-Escoda, C. The use of tricyclic antidepressants in the treatment of temporomandibular joint disorders: Systematic review of the literature of the last 20 years. Med. Oral Patol. Oral Cir. Bucal 2009, 14, E3–E7. [Google Scholar] [PubMed]
  115. Goyal, P.; Singh, R.K.; Gangwar, S.; Mohammad, S.; Pal, U.S.; Singh, G. Effect of duloxetine in temporomandibular joint disorders: A comparison with arthrocentesis. Natl. J. Maxillofac. Surg. 2020, 11, 219–223. [Google Scholar] [CrossRef] [PubMed]
  116. Kimos, P.; Biggs, C.; Mah, J.; Heo, G.; Rashiq, S.; Thie, N.M.; Major, P.W. Analgesic action of gabapentin on chronic pain in the masticatory muscles: A randomized controlled trial. Pain 2007, 127, 151–160. [Google Scholar] [CrossRef] [PubMed]
  117. Harkins, S.; Linford, J.; Cohen, J.; Kramer, T.; Cueva, L. Administration of clonazepam in the treatment of TMD and associated myofascial pain: A double-blind pilot study. J. Craniomandib. Disord. 1991, 5, 179–186. [Google Scholar]
  118. Herman, C.R.; Schiffman, E.L.; Look, J.O.; Rindal, D.B. The effectiveness of adding pharmacologic treatment with clonazepam or cyclobenzaprine to patient education and self-care for the treatment of jaw pain upon awakening: A randomized clinical trial. J. Orofac. Pain 2002, 16, 64–70. [Google Scholar] [PubMed]
  119. Kunjur, J.; Anand, R.; Brennan, P.A.; Ilankovan, V. An audit of 405 temporomandibular joint arthrocentesis with intra-articular morphine infusion. Br. J. Oral Maxillofac. Surg. 2003, 41, 29–31. [Google Scholar] [CrossRef]
  120. List, T.; Tegelberg, Å.; Haraldson, T.; Isacsson, G. Intra-articular morphine as analgesic in temporomandibular joint arthralgia/osteoarthritis. Pain 2001, 94, 275–282. [Google Scholar] [CrossRef]
  121. List, T.; Axelsson, S.; Leijon, G. Pharmacologic interventions in the treatment of temporomandibular disorders; atypical facial pain; and burning mouth syndrome. A qualitative systematic review. J. Orofac. Pain 2003, 17, 301–310. [Google Scholar] [CrossRef]
  122. Gauer, R.L.; Semidey, M.J. Diagnosis and treatment of temporomandibular disorders. Am. Fam. Physician 2015, 91, 378–386. [Google Scholar]
  123. Häggman-Henrikson, B.; Alstergren, P.; Davidson, T.; Högestätt, E.D.; Östlund, P.; Tranaeus, S.; Vitols, S.; List, T. Pharmacological treatment of oro-facial pain-health technology assessment including a systematic review with network meta-analysis. J. Oral Rehabil. 2017, 44, 800–826. [Google Scholar] [CrossRef] [PubMed]
  124. Wu, M.; Cai, J.; Yu, Y.; Hu, S.; Wang, Y.; Wu, M. Therapeutic Agents for the Treatment of Temporomandibular Joint Disorders: Progress and Perspective. Front. Pharmacol. 2020, 11, 596099. [Google Scholar] [CrossRef]
  125. Gil-Martínez, A.; Paris-Alemany, A.; López-de-Uralde-Villanueva, I.; La Touche, R. Management of pain in patients with temporomandibular disorder (TMD): Challenges and solutions. J. Pain Res. 2018, 11, 571–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Kapos, F.P.; Exposto, F.G.; Oyarzo, J.F.; Durham, J. Temporomandibular disorders: A review of current concepts in aetiology, diagnosis and management. Oral Surg. 2020, 13, 321–334. [Google Scholar] [CrossRef] [PubMed]
  127. List, T.; Jensen, R.H. Temporomandibular disorders: Old ideas and new concepts. Cephalalgia 2017, 37, 692–704. [Google Scholar] [CrossRef] [Green Version]
  128. Ratnayake, J.; Guan, G.; Polonowita, A.; Gray, A.R.; Loch, C.; Li, K.C.; Waddell, J.N.; Lyons, K.; Brunton, P.A. Measuring Changes in Jaw Opening Forces to Assess the Degree of Improvement in Patients with Temporomandibular Disorders. Appl. Sci. 2022, 12, 1224. [Google Scholar] [CrossRef]
  129. de Sire, A.; Marotta, N.; Marinaro, C.; Curci, C.; Invernizzi, M.; Ammendolia, A. Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis. Int. J. Mol. Sci. 2021, 22, 5722. [Google Scholar] [CrossRef]
  130. Wieckiewicz, M.; Boening, K.; Wiland, P.; Shiau, Y.Y.; Paradowska-Stolarz, A. Reported concepts for the treatment modalities and pain management of temporomandibular disorders. J. Headache Pain 2015, 16, 106. [Google Scholar] [CrossRef]
  131. Dinsdale, A.; Costin, B.; Dharamdasani, S.; Page, R.; Purs, N.; Treleaven, J. What conservative interventions improve bite function in those with temporomandibular disorders? A systematic review using self-reported and physical measures. J. Oral Rehabil. 2022, 49, 456–475. [Google Scholar] [CrossRef] [PubMed]
  132. Storm Mienna, C.; Glas, L.; Magnusson, M.; Ilgunas, A.; Häggman-Henrikson, B.; Wänman, A. Patients’ experiences of supervised jaw-neck exercise among patients with localized TMD pain or TMD pain associated with generalized pain. Acta Odontol. Scand. 2019, 77, 495–501. [Google Scholar] [CrossRef]
  133. Contento, V.S.; Dalton, B.H.; Power, G.A. The inhibitory tendon-evoked reflex is increased in the torque-enhanced state following active lengthening compared to a purely isometric contraction. Brain Sci. 2019, 10, 13. [Google Scholar] [CrossRef] [Green Version]
  134. Shimada, A.; Ishigaki, S.; Matsuka, Y.; Komiyama, O.; Torisu, T.; Oono, Y.; Sasaki, K. Effects of exercise therapy on painful temporomandibular disorders. J. Oral Rehabil. 2019, 46, 475–481. [Google Scholar] [CrossRef] [PubMed]
  135. Allum, J.H.J.; Gresty, M.; Keshner, E.; Shupert, C. The control of head movements during human balance corrections. J. Vestib. Res. 1997, 7, 189–218. [Google Scholar] [CrossRef] [PubMed]
  136. Xie, Q.; Li, X.; Xu, X. The difficult relationship between occlusal interferences and temporomandibular disorder—Insights from animal and human experimental studies. J. Oral Rehabil. 2013, 40, 279–295. [Google Scholar] [CrossRef]
  137. Alhilou, A.M.; Shimada, A.; Svensson, C.I.; Svensson, P.; Ernberg, M.; Cairns, B.E.; Christidis, N. Nerve growth factor and glutamate increase the density and expression of substance P-containing nerve fibers in healthy human masseter muscles. Sci. Rep. 2021, 11, 15673. [Google Scholar] [CrossRef]
  138. Huang, F.; Zhang, M.; Chen, Y.J.; Li, Q.; Wu, A.Z. Psychological stress induces temporary masticatory muscle mechanical sensitivity in rats. J. Biomed. Biotechnol. 2011, 2011, 720603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Manfredini, D.; Lombardo, L.; Siciliani, G. Temporomandibular disorders and dental occlusion. A systematic review of association studies: End of an era? J. Oral Rehabil. 2017, 44, 908–923. [Google Scholar] [CrossRef]
  140. Ferrillo, M.; Marotta, N.; Giudice, A.; Calafiore, D.; Curci, C.; Fortunato, L.; Ammendolia, A.; de Sire, A. Effects of Occlusal Splints on Spinal Posture in Patients with Temporomandibular Disorders: A Systematic Review. Healthcare 2022, 10, 739. [Google Scholar] [CrossRef] [PubMed]
  141. Mattyasovszky, S.G.; Langendorf, E.K.; Ritz, U.; Schmitz, C.; Schmidtmann, I.; Nowak, T.E.; Wagner, D.; Hofmann, A.; Rommens, P.M.; Drees, P. Exposure to radial extracorporeal shock waves modulates viability and gene expression of human skeletal muscle cells: A controlled in vitro study. J. Orthop. Surg. Res. 2018, 13, 75. [Google Scholar] [CrossRef] [Green Version]
  142. Ammendolia, A.; Marotta, N.; Demeco, A.; Marinaro, C.; Moggio, L.; Barletta, M.; Costantino, C. Effectiveness of radial shockwave therapy in calcific and non-calcific tendinopathy of the shoulder: A systematic review and meta-analysis. Muscles Ligaments Tendons J. 2020, 10, 40–47. [Google Scholar] [CrossRef] [Green Version]
  143. Ramon, S.; Gleitz, M.; Hernandez, L.; Romero, L.D. Update on the efficacy of extracorporeal shockwave treatment for myofascial pain syndrome and fibromyalgia. Int. J. Surg. 2015, 24, 201–206. [Google Scholar] [CrossRef]
  144. Hausdorf, J.; Lemmens, M.A.; Heck, K.D.; Grolms, N.; Korr, H.; Kertschanska, S.; Steinbusch, H.W.; Schmitz, C.; Maier, M. Selective loss of unmyelinated nerve fibers after extracorporeal shockwave application to the musculoskeletal system. Neuroscience 2008, 155, 138–144. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, J.; Huang, Z.; Ge, M.; Gao, M. Efficacy of low-level laser therapy in the treatment of TMDs: A meta-analysis of 14 randomised controlled trials. J. Oral Rehabil. 2015, 42, 291–299. [Google Scholar] [CrossRef] [PubMed]
  146. Tsai, S.-R.; Hamblin, M.R. Biological effects and medical applications of infrared radiation. J. Photochem. Photobiol. B 2017, 170, 197–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Douglas De Oliveira, D.W.; Lages, F.S.; Guimarães, R.C.; Pereira, T.S.; Botelho, A.M.; Glória, J.C.R.; Tavano, K.T.A.; Gonçalves, P.F.; Flecha, O.D. Do TMJ symptoms improve and last across time after treatment with red (660 nm) and infrared (790 nm) low level laser treatment (LLLT)? A survival analysis. Cranio 2017, 35, 372–378. [Google Scholar] [CrossRef] [PubMed]
  148. Ammendolia, A.; Marotta, N.; Marinaro, C.; Demeco, A.; Mondardini, P.; Costantino, C. The synergic use of the High Power Laser Therapy and Glucosamine sulfate in Knee osteoarthritis: A Randomized Controlled Trial. Acta Biomed. 2021, 92, e2021237. [Google Scholar] [PubMed]
  149. Johnson, J.F. Laser therapy and pain management. In Laser Therapy in Veterinary Medicine: Photobiomodulation; Wiley-Blackwell: Ames, IA, USA, 2017; pp. 75–87. [Google Scholar]
  150. Ammendolia, A.; Cespites, M.; Iocco, M. Topical use of aloe gel and low-level laser therapy in overuse tendinitis of elite volleyball players: A randomized controlled trial. Sport Sci. Health 2016, 12, 209–213. [Google Scholar] [CrossRef]
  151. Yasmeen, S. Low Level Laser Therapy Review. Int. J. Curr. Res. 2020, 12, 144. [Google Scholar]
  152. Shanavas, M.; Chatra, L.; Shenai, P.; Rao, P.K.; Jagathish, V.; Kumar, S.P.; Naduvakkattu, B. Transcutaneous electrical nerve stimulation therapy: An adjuvant pain controlling modality in TMD patients—A clinical study. Dent. Res. J. 2014, 11, 676–679. [Google Scholar]
  153. Vance, C.G.; Dailey, D.L.; Rakel, B.A.; Sluka, K.A. Using TENS for pain control: The state of the evidence. Pain Manag. 2014, 4, 197–209. [Google Scholar] [CrossRef]
  154. Bergeron-Vézina, K.; Corriveau, H.; Martel, M.; Harvey, M.P.; Léonard, G. High-and low-frequency transcutaneous electrical nerve stimulation does not reduce experimental pain in elderly individuals. Pain 2015, 156, 2093. [Google Scholar] [CrossRef] [PubMed]
  155. Mummolo, S.; Nota, A.; Tecco, S.; Caruso, S.; Marchetti, E.; Marzo, G.; Cutilli, T. Ultra-low-frequency transcutaneous electric nerve stimulation (ULF-TENS) in subjects with craniofacial pain: A retrospective study. Cranio 2020, 38, 396–401. [Google Scholar] [CrossRef] [PubMed]
  156. Ewall, G.; Parkins, S.; Lin, A.; Jaoui, Y.; Lee, H.K. Cortical and Subcortical Circuits for Cross-Modal Plasticity Induced by Loss of Vision. Front. Neural Circuits 2021, 25, 665009. [Google Scholar] [CrossRef] [PubMed]
  157. Hallman, D.; Lyskov, E. Autonomic Regulation in Musculoskeletal Pain; IntechOpen: London, UK, 2012. [Google Scholar]
  158. Naviaux, R.K. Metabolic features and regulation of the healing cycle—A new model for chronic disease pathogenesis and treatment. Mitochondrion 2019, 46, 278–297. [Google Scholar] [CrossRef]
  159. Zhu, H. Acupoints Initiate the Healing Process. Med. Acupunct. 2014, 26, 264–270. [Google Scholar] [CrossRef] [Green Version]
  160. Vanderploeg, K.; Yi, X. Acupuncture in modern society. J. Acupunct. Meridian Stud. 2009, 2, 26–33. [Google Scholar] [CrossRef] [Green Version]
  161. Kwon, S.; Lee, Y.; Park, H.J.; Hahm, D.H. Coarse needle surface potentiates analgesic effect elicited by acupuncture with twirling manipulation in rats with nociceptive pain. BMC Complement. Altern. Med. 2017, 17, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Wong, L.B. Acupuncture in dentistry: Its possible role and application. Proc. Singap. Healthc. 2012, 21, 48–56. [Google Scholar] [CrossRef]
  163. Cui, J.; Song, W.; Jin, Y.; Xu, H.; Fan, K.; Lin, D.; Hao, Z.; Lin, J. Research Progress on the Mechanism of the Acupuncture Regulating Neuro-Endocrine-Immune Network System. Vet. Sci. 2021, 8, 149. [Google Scholar] [CrossRef]
  164. Park, H.J.; Lee, Y.; Lee, J.; Park, C.; Moon, D.E. The effects of botulinum toxin A on mechanical and cold allodynia in a rat model of neuropathic pain. Can. J. Anaesth. 2006, 53, 470–477. [Google Scholar] [CrossRef]
  165. Baricich, A.; Picelli, A.; Santamato, A.; Carda, S.; de Sire, A.; Smania, N.; Cisari, C.; Invernizzi, M. Safety Profile of High-Dose Botulinum Toxin Type A in Post-Stroke Spasticity Treatment. Clin. Drug Investig. 2018, 38, 991–1000. [Google Scholar] [CrossRef] [PubMed]
  166. Foster, L.; Clapp, L.; Erickson, M.; Jabbari, B. Botulinum toxin A and chronic low back pain. A randomized; double-blind study. Neurology 2001, 56, 1290–1293. [Google Scholar] [CrossRef] [PubMed]
  167. Lippi, L.; de Sire, A.; Folli, A.; D’Abrosca, F.; Grana, E.; Baricich, A.; Carda, S.; Invernizzi, M. Multidimensional Effectiveness of Botulinum Toxin in Neuropathic Pain: A Systematic Review of Randomized Clinical Trials. Toxins 2022, 14, 308. [Google Scholar] [CrossRef] [PubMed]
  168. Bernetti, A.; Agostini, F.; de Sire, A.; Mangone, M.; Tognolo, L.; Di Cesare, A.; Ruiu, P.; Paolucci, T.; Invernizzi, M.; Paoloni, M. Neuropathic Pain and Rehabilitation: A Systematic Review of International Guidelines. Diagnostics 2021, 11, 74. [Google Scholar] [CrossRef] [PubMed]
  169. Muñoz-Lora, V.R.M.; Dugonjić Okroša, A.; Matak, I.; Del Bel Cury, A.A.; Kalinichev, M.; Lacković, Z. Antinociceptive Actions of Botulinum Toxin A1 on Immunogenic Hypersensitivity in Temporomandibular Joint of Rats. Toxins 2022, 14, 161. [Google Scholar] [CrossRef] [PubMed]
  170. Tang, M.; Meng, J.; Wang, J. New Engineered-Botulinum Toxins Inhibit the Release of Pain-Related Mediators. Int. J. Mol. Sci. 2019, 21, 262. [Google Scholar] [CrossRef] [Green Version]
  171. Antoniazzi, C.; Belinskaia, M.; Zurawski, T.; Kaza, S.K.; Dolly, J.O.; Lawrence, G.W. Botulinum Neurotoxin Chimeras Suppress Stimulation by Capsaicin of Rat Trigeminal Sensory Neurons In Vivo and In Vitro. Toxins 2022, 14, 116. [Google Scholar] [CrossRef] [PubMed]
  172. Durham, P.L.; Cady, R.; Cady, R. Regulation of calcitonin gene-related peptide secretion from trigeminal nerve cells by botulinum toxin type A: Implications for migraine therapy. Headache 2004, 44, 35–43. [Google Scholar] [CrossRef]
  173. Teixeira, J.M.; Abdalla, H.B.; Basting, R.T.; Hammock, B.D.; Napimoga, M.H.; Clemente-Napimoga, J.T. Peripheral soluble epoxide hydrolase inhibition reduces hypernociception and inflammation in albumin-induced arthritis in temporomandibular joint of rats. Int. Immunopharmacol. 2020, 87, 106841. [Google Scholar] [CrossRef] [PubMed]
  174. Jîtcă, G.; Ősz, B.E.; Tero-Vescan, A.; Miklos, A.P.; Rusz, C.M.; Bătrînu, M.G.; Vari, C.E. Positive Aspects of Oxidative Stress at Different Levels of the Human Body: A Review. Antioxidants 2022, 11, 572. [Google Scholar] [CrossRef]
  175. Paolucci, T.; Agostini, F.; Bernetti, A.; Paoloni, M.; Mangone, M.; Santilli, V.; Pezzi, L.; Bellomo, R.G.; Saggini, R. Integration of focal vibration and intra-articular oxygen-ozone therapy in rehabilitation of painful knee osteoarthritis. J. Int. Med. Res. 2021, 49, 300060520986705. [Google Scholar] [CrossRef] [PubMed]
  176. Hidalgo-Tallón, F.J.; Torres-Morera, L.M.; Baeza-Noci, J.; Carrillo-Izquierdo, M.D.; Pinto-Bonilla, R. Updated Review on Ozone Therapy in Pain Medicine. Front. Physiol. 2022, 13, 840623. [Google Scholar] [CrossRef] [PubMed]
  177. Bocci, V. The Clinical Application of Ozonetherapy. In OZONE; Springer: Dordrecht, The Netherlands, 2010. [Google Scholar]
  178. Seyam, O.; Smith, N.L.; Reid, I.; Gandhi, J.; Jiang, W.; Khan, S.A. Clinical utility of ozone therapy for musculoskeletal disorders. Med. Gas Res. 2018, 8, 103. [Google Scholar] [PubMed]
  179. Emodi-Perlman, A.; Eli, I.; Smardz, J.; Uziel, N.; Wieckiewicz, G.; Gilon, E.; Grychowska, N.; Wieckiewicz, M. Temporomandibular Disorders and Bruxism Outbreak as a Possible Factor of Orofacial Pain Worsening during the COVID-19 Pandemic-Concomitant Research in Two Countries. J. Clin. Med. 2020, 9, 3250. [Google Scholar] [CrossRef] [PubMed]
  180. Emodi-Perlman, A.; Eli, I. One year into the COVID-19 pandemic–temporomandibular disorders and bruxism: What we have learned and what we can do to improve our manner of treatment. Dent. Med. Probl. 2021, 58, 215–218. [Google Scholar]
  181. Higgins, J.P.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A. (Eds.) Cochrane Handbook for Systematic Reviews of Interventions, Version 6.2; JohnWiley & Sons: Hoboken, NJ, USA, 2021. [Google Scholar]
Figure 1. Pain management and rehabilitation for the central sensitization of temporomandibular disorders.
Figure 1. Pain management and rehabilitation for the central sensitization of temporomandibular disorders.
Ijms 23 12164 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ferrillo, M.; Giudice, A.; Marotta, N.; Fortunato, F.; Di Venere, D.; Ammendolia, A.; Fiore, P.; de Sire, A. Pain Management and Rehabilitation for Central Sensitization in Temporomandibular Disorders: A Comprehensive Review. Int. J. Mol. Sci. 2022, 23, 12164. https://doi.org/10.3390/ijms232012164

AMA Style

Ferrillo M, Giudice A, Marotta N, Fortunato F, Di Venere D, Ammendolia A, Fiore P, de Sire A. Pain Management and Rehabilitation for Central Sensitization in Temporomandibular Disorders: A Comprehensive Review. International Journal of Molecular Sciences. 2022; 23(20):12164. https://doi.org/10.3390/ijms232012164

Chicago/Turabian Style

Ferrillo, Martina, Amerigo Giudice, Nicola Marotta, Francesco Fortunato, Daniela Di Venere, Antonio Ammendolia, Pietro Fiore, and Alessandro de Sire. 2022. "Pain Management and Rehabilitation for Central Sensitization in Temporomandibular Disorders: A Comprehensive Review" International Journal of Molecular Sciences 23, no. 20: 12164. https://doi.org/10.3390/ijms232012164

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop