Exploring the Painless Nature and Potential Mechanisms of Asymptomatic Irreversible Pulpitis: A Narrative Review
REVIEW ARTICLE

Exploring the Painless Nature and Potential Mechanisms of Asymptomatic Irreversible Pulpitis: A Narrative Review

The Open Dentistry Journal 23 Feb 2024 REVIEW ARTICLE DOI: 10.2174/0118742106281444240219050149

Abstract

Background

Considering the extensive innervation of the pulp tissue, asymptomatic irreversible pulpitis (AIP) or “silent pulpitis” represents a confounding clinical condition. Previous studies have attributed the painless nature of AIP to the inhibition of pulpal nociceptors by local endogenous analgesics. However, there is a lack of recent information concerning its painless nature, and paradoxically, patients with dental pain are diagnosed with AIP daily worldwide. In addition, no recent review has explored the potential AIP-related mechanisms.

Objective

This narrative review aims to explore and update the potential mechanisms involved in the painless nature of AIP to improve our current understanding of the asymptomatic character of this clinical condition.

Methods

An electronic search was performed in the PubMed and Scopus databases, using as search terms “asymptomatic irreversible pulpitis,” “dental pulp,” “endogenous opioids,” “endogenous cannabinoids,” “somatostatin,” “GABA,” “bombesin,” “cortistatin,” “galanin,” and “specialized pro-resolving lipid mediators.”

Results

Endogenous opioids, G protein-activated inwardly rectifying K+ channels, endogenous cannabinoids, γ-aminobutyric acid, and neuropeptides (i.e. somatostatin, cortistatin, galanin, and bombesin) could be involved in AIP-related analgesia. Additionally, specialized pro-resolving lipid mediators, such as lipoxins, resolvins, maresins, and protectins, as well as oxytocin, phoenixin, opiorphin, and adipokines, could also be involved in this clinical condition.

Conclusion

This narrative review provides updated information on the potentially involved mechanisms in AIP. Nevertheless, the precise mechanisms responsible for the lack of symptoms in AIP remain to be elucidated, and further research is warranted.

Keywords: Asymptomatic irreversible pulpitis, Analgesia, Endogenous opioids, Endogenous cannabinoids, Neuropeptides, Phoenixin.

1. INTRODUCTION

The dental pulp is a highly vascularized, innervated connective tissue that provides vitality and sensitivity to teeth [1, 2]. This tissue experiences inflammatory reactions in response to dental caries, restorative procedures, dental trauma, and periodontal disease [3] — the first being the main threat to dental pulp [4]. However, pulpitis is the most common inflammatory disease in humans and other mammals [5] that can be reversible or irreversible. Irreversible pulpitis can be symptomatic or asymptomatic [6].

Considering the extensive nerve plexus of the pulp tissue, asymptomatic irreversible pulpitis (AIP) or “silent pulpitis” is a confounding clinical condition [7]. Patients do not experience pain (6) despite inherent inflammatory processes in the affected tooth. Several studies have identified local analgesic agents in the dental pulp, including endogenous opioids [8-10], cannabinoids [11, 12], gamma-aminobutyric acid (GABA) [13, 14], and somatostatin [10, 15]. These agents could inhibit pulpal nociceptors and may be responsible for the asymptomatic nature of AIP [7].

Despite these studies investigating the dental pulp, there is a lack of updated literature reviews that explore the factors involved in the asymptomatic nature of AIP, and the most recent foundational study on the painless nature of AIP dates back two decades [16]. Moreover, patients with dental diseases are diagnosed with AIP on a daily basis worldwide. Therefore, this narrative review aimed to compile updated information on potential factors involved in the mechanisms of analgesia underlying AIP to improve our current understanding of its painless nature and to provide insights for future studies elucidating the precise mechanisms underlying the lack of symptoms in AIP.

2. MATERIAL AND METHODS

We searched the available literature in the PubMed and Scopus databases to identify relevant articles published up to January 25, 2022, describing the expression of ligands and/or receptors or other factors that potentially regulate the asymptomatic nature of AIP. The following search terms were used: “asymptomatic irreversible pulpitis,” “dental pulp,” “endogenous opioids,” “endogenous cannabinoids,” “somatostatin,” “GABA,” “bombesin,” “cortistatin,” “galanin,” and “specialized pro-resolving lipid mediators.” Only articles published in English were included in the present study. The search was limited to clinical trials, in vitro studies, literature reviews, systematic reviews, and textbook chapters. Interim reports, abstracts only, letters, brief communications, studies that did not focus on the asymptomatic nature of AIP, and duplicated works were excluded. Subsequently, the titles and abstracts of relevant articles were reviewed, and a manual search of the references of each selected article was performed to complement the electronic search. Further, endodontic journals were examined to identify relevant articles “in press” or in “early view” status.

3. POTENTIAL FACTORS INVOLVED IN ANALGESIA DURING AIP

Factors that could potentially be involved in the mechanisms of analgesia underlying AIP include endogenous opioids, G protein-activated inwardly rectifying K+ channels (GIRK), endogenous cannabinoids, γ-aminobutyric acid (GABA), neuropeptides (somatostatin, cortistatin, galanin, and bombesin), and specialized pro-resolving lipid mediators (i.e. lipoxins, resolvins, and maresins). We also included a section on miscellaneous factors that could potentially be involved in AIP, such as bacteria and their antinociceptive effects, oxytocin, phoenixin, opiorphin, and adipokines. All but bacteria are endogenous biomolecules potentially involved in the painless nature of AIP. The ligands/receptors of these biomolecules have already been identified in the dental pulp tissue and/or the trigeminal ganglion (TG). However, in some cases, information is not yet available (Table 1).

3.1. Endogenous Opioids

The opioid system functions as an endogenous mechanism of antinociception through three pathways: inhibition of nociceptors at the supraspinal level, inhibition of nociceptors at the level of the dorsal horn of the spinal cord, and activation of the descending inhibitory pathways [15]. This system is distributed in both the central nervous system (CNS) and peripheral nervous system (PNS) [17-20]. It is comprised of endogenous opioid peptides (EOP) (i.e., enkephalins, dynorphins, β-endorphins, and nociceptin/orphanin FQ) [17] released by T- and B-lymphocytes, monocytes, macrophages, and granulocytes [21-24]; and opioid receptors (OR) that are located in the nerve endings of the primary afferent fibers [25, 26]. Met-enkephalins, dynorphins, and β-endorphins have been found in the dental pulp [10, 27, 28]; however, the presence of nociceptin/orphanin has not yet been demonstrated.

The receptors found in the afferent sensory nerves are μ (MOR), δ (DOR), and κ (KOR) [17, 23, 29-32], along with the nociceptin/orphanin receptor (NOR), also known as the orphan opioid receptor-like receptor (ORL) [17, 33]. ORs, especially DOR, are involved in neuroprotection against hypoxia or ischemia [34-37]. They also inhibit voltage-gated Ca2+ channels [38], reduce the release of neurotransmitters [17], and allow neuronal hyperpolarization that is mediated by K+ channels [39]. The presence of MOR [9] and DOR [40] in the dental pulp has been confirmed; however, the KOR expression has not yet been demonstrated.

Under normal conditions, the ORs are inaccessible due to the perineural barrier [41-43]; however, this is altered in initial inflammatory conditions that allow for the passage of ligands to their receptors [30]. In late inflammatory stages, the number of ORs increases, along with their axonal transport to the periphery [9, 24] and sprouting of new nerve endings [44, 45] that would explain the opioid analgesic activity in inflamed dental pulps [9]. In these states, β-endorphins become unstable due to their rapid metabolism; therefore, the resulting analgesia could be induced by their fragments after their biotransformation [46]. In contrast, a previous study showed that antinociception failed to increase despite the increased leukocyte recruitment, which could be attributed to the low amount of ORs in the early inflammatory stage [47]. The duration of inflammation could be a decisive factor in terms of the analgesic capacity of the endogenous opioid system, such that in both types of irreversible pulpitis (symptomatic and asymptomatic), an inflammatory process is established; however, pain is absent in the latter scenario. In endogenous analgesia, the number of leukocytes, ORs, duration of inflammation, and binding efficiency of ORs with G-protein in neurons interact in a simultaneous manner [45, 48, 49].

Central and peripheral ORs interact in the initial inflammatory stages [47, 50-52]; however, in the late stages, only peripheral ORs function [23, 53, 54], demonstrating high participation of peripheral opioid mechanisms as inflammation advances and becomes more severe [48, 51, 55, 56]. Moreover, during chronic inflammation, at the central level, changes in the ORs are not observed, but the levels of EOP increase [32]. Therefore, further studies are required to determine the differences in central responses that occur at different times and in different types of dental injuries [57].

Bradykinin stimulation [58], orthodontic movements [59], and cavity preparation [60] increase the EOP levels in the dental pulp [15, 55, 61]. Moreover, other substances exert an antinociceptive effect as a secondary function by stimulating the release of EOPs in a similar manner to the effects exerted by substance P (SP) whose N-terminal fragment acts as a ligand for MOR [62, 63], calcitonin gene-related peptide (CGRP) that suppresses IL-2 production [64], and IL-4 that promotes change in the phenotype of macrophages from M1 to M2 and stimulates M2 to produce EOP in injured nerves [65]. Additionally, interleukin 1β (IL-1β), corticotropin-releasing factor (CRF) [66], norepinephrine, and CXCL2/3 stimulate the release of EOP by leukocytes [21, 50, 67-72], and thus, exert a peripheral analgesic effect [50, 69-71, 73]. Moreover, some opioid agonists exert anti-inflammatory effects, probably involving ORs on immune cells [74].

In contrast, M2 macrophages can help in resolving inflammatory pain by transferring their mitochondria to the neurons of the dorsal root ganglion (DRG) and stimulating the switch from neuronal glycolytic metabolism to more oxidative metabolism, which in turn regulates the neuronal activity and allows for the resolution of inflammatory pain away from the inflammation site [75]. An increase in the M2 levels has been observed in the TG as pulpitis progresses, showing anti-inflammatory effects [76]. Moreover, this analgesic effect could be attributed to the secretion of IL-10 because of its anti-inflammatory action [77-81] and a reduction in the expression of voltage-gated sodium channels and a number of currents sensitive to tetrodotoxin [82]. However, the resolution of inflammation is insufficient to resolve the pain [80].

3.2. GIRK

GIRK are G protein-activated effector ion channels [83] that participate in opioid-mediated antinociception in the CNS and PNS via hyperpolarization of the neuronal membrane, which in turn inhibits the propagation of action potentials [84-90]. At the spinal cord level, these receptors contribute to the analgesic effects of MOR and DOR but not those of KOR [88]. Furthermore, GIRK channels are crucial for galanin action, as GalR1 and GalR3 open the K+ channels. For neuropeptide Y, which presynaptically depresses the miniature excitatory synaptic currents through the Y2 receptor, somatostatin activates the GIRK channels of SST4 receptors [91-95].

The GIRK 1 and 2 receptors are expressed in the TG neurons, thus contributing to peripheral opioid analgesia in the craniofacial region [96]. Therefore, these channels could be present in the dental pulp; however, to date, no study has confirmed this hypothesis.

3.3. Endogenous Cannabinoids

The endogenous opioid and cannabinoid systems are involved in antinociception through different pathways [97-104]. Moreover, they activate the G-protein-coupled receptors (GPCR) and can interact either directly (receptor heteromerization) or indirectly (cross-signaling) [97, 98, 105]. Moreover, cannabinoid receptors (CBRs) activate GIRK, which in turn reduces the release of neurotransmitters in the opioid system [106]. The endocannabinoid system has receptors (CB1R and CB2R), endogenous ligands (anandamide and 2-arachidonylglycerol), and enzymes that degrade and synthesize the latter, performing functions at the central and peripheral levels [107, 108]. This system is expressed in both the ascending and descending pain pathways, producing antinociception at the supraspinal, spinal, and peripheral levels [102, 109-112]. Additionally, lipopolysaccharides (LPS) increase the levels of anandamide and inhibit the enzyme fatty acid amide hydrolase (FAAH) in the lymphocytes [113], and increase the levels of 2-arachidonylglycerol (2-AG) in the macrophages and platelets [114].

CB1R and CB2R are mainly expressed in the nervous and immune systems, respectively [115-119], and the cells of these systems secrete endocannabinoids [23, 120, 121]. CB1Rs have been identified in various areas related to pain in the CNS, where they regulate signals from neurons originating from the nociceptive regions of the spinal cord, producing antinociception [110-112, 118]. CB1R of the ventrolateral periaqueductal gray matter (vlPAG) aids in modulating the nociceptive signals from the TG nerve, specifically in capsaicin-induced pulpal pain [122]. The exact mechanism of this modulation is unclear; however, CB1R in the PAG interacts with other systems to modulate the nociceptive signals [123, 124], such as orexin 1 receptors (OX1Rs). When activated, these receptors induce the release of 2-AG, which inhibits the release of GABA through the pre-synaptic CB1R—a phenomenon known as disinhibition [123]. Tonic inhibition of GABAergic transmission activates the vlPAG; thus, activating the descending pain inhibition pathway [125]. This demonstrates the antinociceptive effects of orexin-A on the vlPAG and its relationship with the endocannabinoid system [123].

The CBR signaling pathway acts through the inhibition of cyclic-AMP formation and modulation of Ca2+ and K+ channels [126]. Different ligands differentially activate these signaling pathways through CB1R and CB2R— which is termed the “biased signaling” [127]. Further studies at the pulp level are suggested as this signaling may preferentially provide higher analgesia. Additionally, the molecular mechanisms underlying the antinociceptive and antihyperalgesic effects of CBRs remain unclear [115]. Endocannabinoids, such as anandamide, 2-AG, and the other less-studied subtypes, including N-arachidonoyl-dopamine (NADA), noladin ether, and virodhamine, interact with receptors other than CB1R and CB2R [105, 128, 129].

In contrast, transient receptor potential vanilloid subtype 1 channels (TRPV1) are activated by anandamide and NADA and are co-expressed with CB1R and CB2R in some tissues, including the dental pulp [128, 130]. This co-expression or “cross-talk” between CBR and TRPV1 may be relevant in pulpal analgesia. It has long been known that only CBRs attenuate and TRPV1 increases nociception. However, studies have shown that TRPV1 activation potentiates the supraspinal pain inhibitory pathways, and desensitization of TRPV1 produces analgesia [131, 132]. Pre- and post-synaptic activation of TRPV1 or pre-synaptic activation of CB1R stimulates the output excitatory neurons through glutamate release or disinhibition of GABA tonic control, respectively, at the vlPAG level. This leads to glutamate release in the rostral ventromedial medulla (RVM) and activation of the “off” neurons in this area, with a subsequent antinociception [133, 134]. However, further studies are required to analyze the factors that activate these pathways.

TRP channels also induce peripheral antihyperalgesia and antinociception [135, 136]. However, their mechanism of action is complex, as they generate incoming ionic currents more associated with nociception. Partial activation of these channels may not necessarily generate neuronal excitation [137-139]. The incoming currents could fail to reach threshold levels to excite the nociceptors, or the slow depolarization of the membrane potential may inactivate these channels [140].

CB1R and CB2R expression in the dental pulp of humans and rats has been previously demonstrated [10, 11, 130, 141-143]. CB2R is expressed in the human pulp cells [130, 142] and myofascial fibroblasts [144], whereas CB1R is preferentially expressed in odontoblasts, odontoblast-like cells, and pulpal nerve fibers [11, 145]. Although the expression of CBRs has not been shown in dental pulp fibroblasts, it has been reported that fibroblasts have the necessary enzymes to produce endocannabinoids and act in an autocrine or paracrine way when interacting with leukocytes [144, 146, 147]. A previous study showed that there were no statistically significant differences in the expression of CB1R between painful and non-painful dental pulps [11].

In contrast, CB1R may be activated by stretching in the absence of a ligand [119], wherein hydrostatic pressure may directly activate OR and CBR, releasing endorphins and endocannabinoids, as has been reported at the PAG level [148]. In the dental pulp, an increase in pressure during an inflammatory process may activate these receptors, although this remains to be explored. However, the role of CB2Rs and their agonists has been investigated in pulpal antinociception [149] and in animal models of acute, chronic, and neuropathic pain [150].

CB2R agonists can inhibit inflammatory pain through their anti-inflammatory effects [151, 152]. Moreover, the expression of cytokines and CBR have a reciprocal regulatory relationship. Thus, the activation of these receptors in macrophages inhibits the production of proinflammatory cytokines [153], thus allowing the change from immune responses mediated by Th1 (proinflammatory) to Th2 (anti-inflammatory) through the CB2R [154]. Moreover, IL-4 increases the CB1R expression in leukocytes [155], and IFN-γ and IL-12 reduce the FAAH activity [113]. Taken together, the increase in CBR expression by cytokines could be a mechanism of autoregulation of inflammation [156].

3.4. GABA

GABA neurotransmitter plays a primary inhibitory role in the CNS and PNS [157-161]. When released at the neuronal synapses, it activates different classes of receptors or returns to the nerve terminals via a Na+-dependent transporter [158-160]. Ionotropic receptors (GABAA and GABAC) participate in rapid synaptic transmission and modulate neuronal activity by gating the chloride ions [13, 159], hyperpolarizing the neuronal membranes, and inhibiting the propagation of action potentials, leading to short-term, fast-acting inhibitory currents [157, 160]. In contrast, the slow-acting metabotropic receptors (GABAB) belong to the GPCR superfamily and exert inhibitory actions through the inhibition of voltage-gated Ca2+ channels and GIRK activation [162-167]. These receptors have been found in the dental pulp tissue [13] and TG [163]. Furthermore, GABAergic neurons are activated at the trigeminal nuclear complex during tooth pulp stimulation [161].

However, inflammation, necrosis, or areas of pulpal hypoxia can increase the GABA levels above the nominal levels at rest, which may explain the absence of symptoms in these pulps [12, 13]. Neuroinflammation can be modulated by GABAergic signaling [157], as GABAB receptors are involved in pain management and analgesia; thus, GABA and GABAB receptors present in the human pulp may also be involved [164]. The clinical importance of peripheral GABAB receptors may be related to the peripheral analgesic effects of GABAB agonists that modulate or attenuated nociceptive behavior in the animal models of pain [165]. In a previous study [166], isovaline, baclofen, and GABA attenuated allodynia induced by prostaglandin E2 injection. Another study revealed that baclofen suppressed pain in small-diameter TG neurons in rats [162].

In contrast, GABAergic interneurons mediate the endogenous release of 5-hydroxytryptamine (5-HT). The 5-HT3 receptors are involved in antinociceptive effects [167] that are attenuated by the opioid antagonist naloxone, suggesting that these neurons may be associated with endogenous opioids [168].

3.5. Neuropeptides: Somatostatin, Cortistatin, Galanin, and Bombesin

Neuropeptides play a major role in the perception of pain [169], but some can mediate analgesic mechanisms [170]. In this section, we describe the potential analgesic roles of somatostatin, cortistatin, galanin, and bombesin in AIP.

3.5.1. Somatostatin (SST)

SST is a peptide hormone [171-173] that is widely distributed in the CNS and peripheral tissues [171, 174] and is produced by neurons and neuroendocrine, inflammatory, and immune cells in response to ions, nutrients, neuropeptides, neurotransmitters, hormones, growth factors, cytokines [173], and noxious heat or chemical stimuli [175]. There are two SST isoforms, SST-14 and SST-28, that differ in the number of amino acids [175-177] and five GPCR-type receptors (SSTR 1–5) [178].

SST performs antinociceptive functions [179-182] by affecting neurotransmission through its receptors, decreasing the conductance of voltage-gated Ca2+ channels [172, 183], and activating K+ channels [184-186]. SST decreases neurogenic inflammation [175] due to its inhibitory action [175, 187, 188] by decreasing the release of IFN-γ, reactive oxygen species, CGRP [175], SP [189], and immunoglobulins from B-cells [190]. Moreover, SST can regulate the pulpal blood flow [191] as the peptidergic nerves containing SST are distributed near the blood vessels [10, 192-194].

3.5.2. Cortistatin (CORT)

CORT, a cyclic neuropeptide, is predominantly expressed in the cerebral cortex [195-197], spinal cord neurons, GABAergic inhibitory interneurons [198-200], immune cells (lymphocytes, monocytes, macrophages, and dendritic cells) [201, 202], and to a lesser extent in endothelial cells, endocrine cells, peripheral nociceptive neurons, and smooth muscle cells [203] in response to noxious stimuli, cytokines, and tissue injury [197, 203].

CORT binds with a high affinity to different receptors, mainly SSTR 1–5 [192, 204], ghrelin receptor (GHSR1) [196, 197], and an unidentified selective CORT receptor [198]. It shares several functions with SST [205], such as suppression of nerve function and inhibition of cell proliferation [196, 206]; however, it has other functions, such as sleep induction, reduction of locomotor activity, and deactivation of inflammatory/autoimmune responses [196, 203, 206, 207]. Regarding adaptive immunity, CORT acts on CD4 T-lymphocytes, participates in the inhibition of differentiation and activation of Th1 and Th17 lymphocytes, and induces differentiation and activation of Th2 and Treg lymphocytes. As for innate immunity, CORT acts on macrophages/monocytes and participates in the inhibition of proinflammatory mediators, such as CGRP [205], TNF, IL-6, IL-12, IL-1, NO, GM-CSF, and CK, and increases the levels of IL-10 [208, 209]; thus, exerting anti-inflammatory effects. In contrast, its deficiency can exacerbate inflammatory pain responses [197, 210].

Finally, CORT is capable of deactivating microglia and astrocytes in an inflammatory environment [197, 211]. Activated glial cells play a critical role in the development and maintenance of nociceptive responses, especially at the spinal cord level [211]. Thus, CORT regulates inflammation-induced pain through deactivation, particularly by preventing the development of chronic pain. It also relieves hyperalgesia and allodynia and acts as a neuroprotector and neuroregenerator [205]. Furthermore, CORT mRNA and protein are detected in mature and newly developing odontoblasts. Thus, SSTR1 and CORT may have important functions in the regulation of pulpal inflammation and communication between odontoblasts and the nervous system [212] and may be involved in antinociceptive processes at the pulpal level. However, further studies are needed to confirm these hypotheses in the dental pulp.

3.5.3. Galanin (GAL)

GAL, a neuropeptide widely distributed in the CNS and PNS [213-216], is present in non-neuronal cells, such as keratinocytes, sweat glands, macrophages, and blood vessels [217]. It is expressed by immune cells during inflammation in an attempt to restore homeostasis [218] and exerts its physiological effects through three types of GPCRs [216, 219], namely GalR1, GalR2, and GalR3 [216, 220-224]. Previous studies have suggested that GAL and its receptors may be involved in the transmission and modulation of nociceptive information in the nervous system [225-230].

GAL has an antinociceptive effect [228, 231-235] via activation of GalR1 [226, 236-238] and GalR3, which causes neuronal hyperpolarization in response to increased K+ conductance [239], and also favors the release of enkephalins and endorphins in the primary afferent neurons that innervate the dental pulp [240]. The immunoreactivity of GalR1 has been observed in the axoplasm of unmyelinated nerve fibers (type C and Aδ) of the dental pulp [241, 243]. However, it can induce pronociceptive effects [224, 243] through the action of GalR2 [226, 236-238, 244-246] and activation of phospholipase C–protein kinase C pathway [247]. Nevertheless, the GAL action differs according to its concentration, where the activation of GalR2 changes from a Gq pathway (low GAL concentration) to a Gi/o-dependent pathway (high GAL concentration); therefore, it changes from a pro- to an antinociceptive-type signaling pathway [248]. However, the latter has not yet been observed in the dental pulp tissue.

3.5.4. Bombesin (BN)

The endogenous peptide, BN [249], and its homologues, neuromedin B (NMB) and gastrin-releasing peptide (GRP) are important neuromodulators in the brain [250, 251]. They function through three subtypes of G protein-coupled hepta-helical receptors, namely BB1, BB2, and BB3. NMB and GRP show high affinity and serve as endogenous ligands for BB1 and BB2 receptors, respectively [250], whereas BN activates both receptors [251], and BB3 is an orphan receptor with low affinity for all these peptides.

BN increases the presynaptic release of GABA by facilitating the entry of extracellular Ca2+ [250], depolarizes GABAergic interneurons at the presynaptic level through the inhibition of KIRs and K+ conductance, and increases the input resistance of interneurons. This suggests that BN reduces the conductance of the neuronal membrane [250]. Its antinociceptive action may be related to the release of GABAergic interneurons.

A previous study [13] demonstrated the significantly higher presence of specific GABA-like and BN/GRP-like immunoreactivity in the pulps of asymptomatic carious teeth than in normal teeth. Both peptides have been implicated in antinociception [13] and have been reported in TG neurons [252]. Their immunoreactivity has been observed within the pulpal nerves and pulp fibroblasts [13].

3.6. Specialized Pro-resolving Lipid Mediators (SPMs): Lipoxins, Resolvins, Maresins, and Protectins

The SPMs actively resolve inflammation to avoid the development of a chronic condition [253]. These endogenous lipid mediators act as immune response modifiers and selectively modulate and reduce the host response. They resolve inflammation [254] by clearing debris and infectious agents, reducing pain, and restoring the function of damaged tissues [255].

In contrast, several studies support the potent role of SPMs in reducing the different types of pain, including inflammatory and neuropathic pain [256-263], through GPCRs and different downstream mechanisms, such as the regulation of inflammatory mediators, TRP channels, and central sensitization [264].

Studies with animal models indicate that SPMs can reduce inflammatory, postoperative, and neuropathic pain via immune, glial, and neuronal modulation [265]. Additionally, SPMs are produced in small amounts in vivo (nano- or picograms), and thus, the doses used in experimental studies are of equal magnitude [262, 266, 267]. Despite the low doses, the analgesic and anti-inflammatory potency of SPMs is evident. Those doses are not comparable with the milligrams or grams used with analgesic agents, such as nonsteroidal anti-inflammatory drugs or opioids [268, 269].

Furthermore, SPMs could potentially participate in the asymptomatic nature of AIP via the resolution of inflammation and their anti-inflammatory effects. Nevertheless, such issues need to be clarified by well-established pulpal pain models. However, technical barriers pertaining to the instability, complex and delicate physicochemical nature, and metabolic inactivation of SPMs must be overcome [253].

3.7. Miscellaneous Mechanisms

3.7.1. Bacteria and their Antinociceptive Effects

Inflammation-induced pain is an adaptive response designed to protect the body from further injuries [270]. However, disease scenarios vary because some pathogens can block, reduce, or modulate pain during the disease cycle [271]. For instance, Porphyromonas gingivalis [270] is associated with destructive periodontal disease [270, 272], dental caries [273], endodontic infections, and odontogenic abscesses [274]. It exerts antinociceptive effects [270], where its LPS increases the levels of the potent anti-inflammatory cytokine IL-10 [270] and stimulates the peptide derived from human telomerase (GV1001) that has an anti-inflammatory effect without affecting the cell viability in the human dental pulp, as it allows for downregulating the expression of TNF-α and IL-6 [272]. However, the antinociceptive role of this bacterium has not yet been studied in AIP; thus, more studies are required.

Metagenomic studies have revealed that the human microbiome can generate many bioactive molecules, including histamine, epinephrine, and GABA [275-277]. Therefore, the possible antinociceptive actions of bacteria, such as Lactobacillus species [278, 279] (Lactobacillus acidophilus NCFM), induce the expression of the cannabinoid and μ-opioid receptor in the intestinal epithelial cells [280]. Whereas, Bifidobacterium species, such as B. dentium, also produce GABA [281-283], making neurons less likely to reach the threshold depolarization level [283].

In contrast, M. ulcerans can secrete mycobacterial polyketide mycolactone to induce analgesia by activating angiotensin II type 2 receptors (AT2R) and inducing hyperpolarization through activation of K+ channels in nociceptors [271, 284]. Additionally, in acute staphylococcal infections [285], CGRP, GAL, and somatostatin can suppress TNF-α release from S. aureus-stimulated or heat-killed lipoteichoic acid macrophages. This indicates that the presence of these bacterial agents may induce the production of other substances that reduce inflammation and have analgesic action.

Finally, in the dental pulp, LPS from bacteria modulates the nociceptive activity through TLR4-mediated sensitization of TRPV1 to nociceptors [286]. Moreover, LPS could be detrimental if pathogenic factors suppress nociception because they can evade host detection and allow for the silent spread of infection.

3.7.2. Oxytocin (OXT)

OXT, a hormone and neuropeptide, induces antinociception [287-290] and participates in the endogenous opioid system [288]. At the TG level, the expression of OXT receptors (OXTR) in the nociceptive neurons (small A–δ fibers) has been confirmed [289, 290], and their expression increases during chronic inflammation [291]. Both OXT and vasopressin (V1A) and their associated receptors, namely OXTR and V1AR, respectively, induce analgesia in the sensory neurons [292-295], possibly because the peripheral antinociceptive action of vasopressin is due to an increase in the function of the GABAA receptor, inhibition of the acid-sensitive ion channels [293, 296], and OXT by the direct desensitization of TRPV1 [297]. Therefore, the analgesic action may also be present in the dental pulp due to its expression in the TG. However, this requires further investigation.

3.7.3. Phoenixin

The neuropeptide, phoenixin, is expressed in the TG sensory neurons that may not be associated with antinociception in thermal pain models. However, phoenixin is associated with antinociception in the visceral pain models [298]. Phoenixin suppresses LPS-induced inflammation in the dental pulp cells, and its anti-inflammatory effects have been demonstrated by confirming the expression of its receptor, GPR173, in the human pulp cells [299]. Further studies should address its anti-inflammatory and potential analgesic properties.

3.7.4. Opiorphin

Enkephalins have a stronger analgesic effect than morphine, but this effect does not last because of the degrading enzymes, such as neutral endopeptidase and aminopeptidase-N [300]. Opiorphin is a peptide that acts as an inhibitor of these enzymes, thus prolonging the effects of enkephalins [300-302].

It is present in the blood, urine, semen, milk, tears, and saliva, although its highest concentrations have been observed in tears and saliva [303]. The more intense the pain due to inflammation is, the more the salivary opiorphin exists; however, its expression remains to be evaluated in the pulp tissue.

3.7.5. Adipokines

Adipokines play multiple physiological and pathological functions in the dental pulp, and some of them exert anti-inflammatory activity, such as adiponectin and ghrelin [304]; therefore, both adipokines could reduce pain in AIP due to their inherent anti-inflammatory activity. Although several adipokines have recently been identified [305], only a few of them have been studied in the pulp tissue [304]. Thus, their potential involvement in pulp inflammation and pain warrants further investigation.

Table 1 summarizes all the aforementioned factors that are potentially involved in AIP, their ligands/receptors identified in the dental pulp tissue and/or TG, and their potential analgesic-related mechanisms.

Table 1.
Potential factors involved in AIP.
Potential Mechanism Ligands Identified in the Dental Pulp or TG Receptors Identified in the Dental Pulp or TG Role Confirmed in AIP-related Analgesia Potential Mechanisms Involved in the Painless Nature of AIP
Endogenous opioids Dental pulp [8, 10, 15, 27, 28, 58-60]. Dental pulp [9, 40]. No - Negative regulation of neurogenic inflammation [8].
- ORs up-regulation in late inflammatory stages, along with their axonal transport to the periphery [9, 24, 32, 57].
- Peripheral analgesia by the up-regulated expression of ligands and/or receptors [30-32].
- Anti-inflammatory effects [74].
- Pain modulation within the inflamed tissue by opioid peptides released from the immune cells [21-24, 47, 48, 51, 52, 67-72, 80].
- High involvement of the peripheral opioid mechanisms as inflammation advances [48, 51, 55, 56].
GIRK N/A TG [96]. No Peripheral opioid-mediated analgesia [85, 87-89, 96].
Endogenous cannabinoids N/A Dental pulp [10, 11, 130, 141-145, 149]. No - Anti-inflammatory and analgesic effects [136, 145].
- GIRK activation reduces the release of neurotransmitters [106].
- Increased analgesia through biased signaling [127].
- CB1 inhibits the neurotransmitter release on nerve terminals and CB2 modulates cytokine release on immune cells [130].
- Inhibition of inflammatory pain by anti-inflammatory effects [151-154].
- Cross-talk between CBR and TRPV1 may provide pulpal analgesia [130, 132].
GABA Dental pulp [11]. Dental pulp [12, 164].
TG [163].
No - GABA-mediated inhibitory neurotransmission [11, 12, 158, 159].
- 5-HT mediated GABAergic inhibition [167, 168].
- Hyperalgesia reduction by GABA peripheral analgesic effects [165].
- Blood flow regulation through inhibition of noradrenaline release in dental pulp [160].
Neuropeptides
Somatostatin Dental pulp [10]. Dental pulp [212]. No - Anti-inflammatory neuropeptide that down-modulates a number of immune functions [175, 193].
- Decreases the neurogenic inflammation [175, 193].
- Inhibits CGRP release from the trigeminal neurons [205].
- Exerts antinociceptive functions [179-182, 187, 188].
- Decreases the conductance of voltage-gated Ca2+ channels [172, 183] and activates K+ channels [184-186].
Cortistatin Dental pulp [212]. N/A No - Potent anti-inflammatory effect [195, 204, 211] by regulating immune tolerance 2009].
- Deactivation of inflammatory responses [196, 203, 206, 207].
- Decreases the presence/activation of Th1 and Th17 cells in the periphery [211].
- Inhibits pro-inflammatory mediators (TNF, IL-6, IL-12, IL-1, NO, and GM-CSF) and increases the levels of IL-10 [208, 209].
- Relieves hyperalgesia and allodynia and acts as a neuroprotector and neuroregenerator [205].
- Analgesic effect in inflammatory [197] and neuropathic pain [210].
- Inhibits the CGRP release from the trigeminal neurons [205].
- Depresses the neuronal electrical activity [206].- Relieves hyperalgesia and allodynia and acts as a neuroprotector and neuroregenerator [205].
- Analgesic effect in inflammatory [197] and neuropathic pain [210].
- Inhibits the CGRP release from the trigeminal neurons [205].
- Depresses the neuronal electrical activity [206].
Galanin Dental pulp [212].
TG [245].
Dental pulp [241].
TG [241, 245].
No - Antinociceptive effect [225-235, 240].
- Opioid systems are involved in the galanin-induced antinociception [240].
Bombesin Dental pulp [13].
TG [252].
N/A No - Antinociceptive effect [13].
- Depolarizes GABAergic interneurons at the presynaptic level and reduces the conductance of the neuronal membrane [250].
Specialized pro-resolving lipid mediators (SPMs)
Lipoxins N/A N/A No - Potent pro-resolving and anti-inflammatory effects and analgesic action [264, 265].
Resolvins N/A N/A No - Potent pro-resolving and anti-inflammatory effects and analgesic action [264, 265].
- Analgesic effect in inflammatory pain [259, 260, 262, 269].
- Potent inhibition of TRP channels [261].
Maresins N/A N/A No - Potent pro-resolving and anti-inflammatory effects and analgesic action [264, 265].
Protectins N/A N/A No - Potent pro-resolving and anti-inflammatory effects and analgesic action [264, 265].
Miscellaneous mechanisms
Antinociceptive bacteria - - No -Porphyromonas gingivalis LPS exerts antinociceptive effects via an increase in IL-10 levels [270].
-Bifidobacterium species, such as B. dentium, produce GABA [281-283].
Oxytocin TG [290]. TG [290-292]. No - Induces membrane hyperpolarization in pain-sensitive dorsal root ganglia neurons [287].
- Antinociceptive effect [287-290].
- Inhibits the activity of acid-sensing ion channels [293].
- Suppresses nociception of inflammatory pain via TRPV1-desensitization [297].
Phoenixin TG [298]. Dental pulp [299]. No - Suppresses the lipopolysaccharide-induced inflammation in dental pulp cells, suppressing the release of pro-inflammatory cytokines and inflammatory mediators [299].
Opiorphin N/A N/A No - Protects enkephalins from degradation and activates restricted opioid pathways specifically involved in pain control [300-303].
Adipokines Dental pulp [304]. Dental pulp [304]. No Some exert anti-inflammatory
effects by inducing the secretion of
anti-inflammatory interleukins or inhibiting the production of proinflammatory
cytokines [304].
N/A: not available information.

CONCLUSION

This review presents up-to-date information on the painless nature of AIP. Factors that could potentially be involved in the mechanisms of analgesia underlying AIP include endogenous opioids, GIRK channels, endogenous cannabinoids, GABA, neuropeptides (somatostatin, cortistatin, galanin, and bombesin), and SPMs (lipoxins, resolvins, and maresins). We have also identified some miscellaneous factors that could play a role in AIP, such as bacteria with their antinociceptive effects, oxytocin, phoenixin, opiorphin, and adipokines, considering their potential analgesic-related mechanisms.

Nevertheless, the precise mechanisms responsible for the lack of symptoms in AIP remain to be elucidated, and further research is warranted despite the recent advances in science and technology. The available literature mainly investigated symptomatic irreversible pulpitis (SIP), where a recent study determined the levels of inflammation, oxidative stress, and extracellular matrix degradation biomarkers in SIP [305, 306]. Thus, it is compelling to perform a similar biochemical mapping for AIP that helps elucidate the expression pattern of endogenous analgesic biomolecules. Furthermore, vascular, neural, cellular, and biochemical changes can occur without pain (8).

Moreover, it is important to highlight the chronic nature of AIP. In this regard, systemic chronic inflammation constitutes a health-damaging phenotype that is triggered by damage-associated molecular patterns, is persistent (non-resolving), has low-grade magnitude, leads to collateral damage, is age-related, and is silent (has no canonical standard biomarkers) [307]. The influence of these factors should be investigated to collect data concerning the analgesic features and pathophysiology of AIP in the context of the local microenvironment of the pulp tissue.

Furthermore, multiplatform data-integration models have been used to identify the differentially expressed genes to analyze the molecular mechanisms underlying pulpitis [308, 309]. Thus, they could improve our current understanding of the nature of AIP. Genetic and epigenetic characterization of pulpal inflammation can also help decipher the balance between proinflammatory and anti-inflammatory gene expression in AIP [310] and how it influences analgesia. This is especially relevant as several genes known to modulate pain and inflammation show a higher level of differential expression in patients with asymptomatic and mild pain compared to those with moderate to severe pain [311].

Regarding the limitations of this review, it must be highlighted that most studies were performed on animals, and AIP could not be differentiated from SIP. Furthermore, our search was confined to two electronic databases, potentially limiting the inclusion of relevant literature in our review. Despite these limitations, our study possesses notable strengths. We have meticulously compiled a substantial amount of data, contributing to an updated narrative review that delves into the potential mechanisms behind the asymptomatic nature of AIP. Notably, the latest report on the fundamentals of the painless nature of AIP describes a clinical study that was performed two decades ago [16]. Additionally, our findings could offer valuable insights for designing new studies aimed at identifying the precise molecular mechanisms responsible for the absence of symptoms in AIP.

Although the present review enlists some candidate ligands and/or receptors that potentially regulate the asymptomatic nature of AIP, no direct evidence supports these statements (Table 1). Indeed, the literature regarding this topic is scarce. However, paradoxically, patients with dental diseases are diagnosed with AIP daily worldwide. Therefore, understanding the analgesia and biology behind AIP is necessary and could help improve the clinical diagnosis of pulp pathology, especially since recent investigations have shown a good correlation between the clinical symptoms of pulpitis and histological findings [312]. On the other hand, anecdotal reports among dentists confirm that in some AIP cases that may have had trauma or deep caries, the inflamed pulp tissue is open to the oral cavity. This would mean no or little increase in pulpal tissue pressure is induced, which is thought to be involved in the “asymptomatic” AIP condition. However, this assumption is very simplistic in explaining the potential biological fundamentals behind AIP.

Finally, other dental and medical pathologies share asymptomatic characteristics similar to those of AIP. These include symptomless pericoronitis [313, 314], chronic periodontitis [315], asymptomatic apical periodontitis [316-318], congenital painlessness disorders [319-321], painless neuropathies [322], NGF mutations [323], Buruli ulcer [324], and painless chronic pancreatitis [325-327]. Hence, analyzing the cellular, biochemical, and/or clinical findings from these conditions could help enhance our understanding of the possible mechanisms underlying the asymptomatic nature of AIP.

LIST OF ABBREVIATIONS

BN = Bombesin
CBRs = Cannabinoid Receptors
CORT = Cortistatin
EOP = Endogenous Opioid Peptides
FAAH = Fatty Acid Amide Hydrolase
GABA = Gamma-aminobutyric Acid
GAL = Galanin
GIRK = G Protein-activated Inwardly Rectifying K+ Channels
NADA = N-arachidonoyl-dopamine
NOR = Nociceptin/orphanin Receptor
OXT = Oxytocin
OX1Rs = Orexin 1 Receptors
SPMs = Specialized Pro-resolving Lipid Mediators
SST = Somatostatin
TRPV1 = Transient Receptor Potential Vanilloid Subtype 1 Channel
vlPAG = Ventrolateral Periaqueductal Gray Matter
2-AG = 2-arachidonylglycerol

AUTHORS’ CONTRIBUTIONS

D.P.C., V.C.J. and J.L.A. contributed to literature search, writing-original draft preparation, writing–review, and editing. J.L.A. contributed to conceptualization, methodology, critical revision of the article, supervision and project administration. All authors have read and agreed to the published version of the manuscript.

CONSENT FOR PUBLICATION

Not applicable.

AVAILABILITY OF DATA AND MATERIALS

The data and supportive information are available within the article.

FUNDING

None.

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

This article is based on Daniela Paola Cabrera-Abad and Verónica Cristina Jara-Vergara undergraduate thesis.

The APC was supported by the Vice Chancellor's Office for Research of the University of Cuenca (VIUC).

REFERENCES

1
Yu C, Abbott PV. An overview of the dental pulp: its functions and responses to injury. Aust Dent J 2007; 52(1)(Suppl.): S4-S16.
2
Ravindran S, Huang CC, George A. Extracellular matrix of dental pulp stem cells: applications in pulp tissue engineering using somatic MSCs. Front Physiol 2014; 4: 395.
3
Galler KM, Weber M, Korkmaz Y, Widbiller M, Feuerer M. Inflammatory Response Mechanisms of the Dentine–Pulp Complex and the Periapical Tissues. Int J Mol Sci 2021; 22(3): 1480.
4
Kassebaum NJ, Bernabé E, Dahiya M, Bhandari B, Murray CJL, Marcenes W. Global burden of untreated caries: a systematic review and metaregression. J Dent Res 2015; 94(5): 650-8.
5
Wang Y, Zhai S, Wang H, et al. Absent in Melanoma 2 (AIM2) in Rat Dental Pulp Mediates the Inflammatory Response during Pulpitis. J Endod 2013; 39(11): 1390-4.
6
Glickman GN. AAE Consensus Conference on Diagnostic Terminology: background and perspectives. J Endod 2009; 35(12): 1619-20.
7
Byers M, Henry M, Närhi M. Dental innervation and its responses to tooth injury.Seltzer and Bender’s Dental pulp 2012; 133-58.
8
Chavarria-Bolaños D, Flores-Reyes H, Lombana-Sanchez N, Cerda-Cristerna B, Pozos-Guillen A. Sensory neuropeptides and endogenous opioids expression in human dental pulp with asymptomatic inflammation: in vivo study. Mediators Inflamm 2015; 2015879126
9
Jaber L, Swaim WD, Dionne RA. Immunohistochemical localization of mu-opioid receptors in human dental pulp. J Endod 2003; 29(2): 108-10.
10
Casasco A, Calligaro A, Casasco M, et al. Peptidergic nerves in human dental pulp. An immunocytochemical study. Histochemistry 1990; 95(2): 115-21.
11
Mitrirattanakul S, Poomsawat S, Fuangtharnthip P. Cannabinoid receptor 1 (CB1R) expression in rat dental pulp. Oral Sci Int 2012; 9(1): 17-20.
12
Beneng K, Renton T, Yilmaz Z, Yiangou Y, Anand P. Cannabinoid receptor CB1-immunoreactive nerve fibres in painful and non-painful human tooth pulp. J Clin Neurosci 2010; 17(11): 1476-9.
13
Todd WM, Kafrawy AH, Newton CW, Brown CE. Immunohistochemical study of gamma-aminobutyric acid and bombesin/gastrin releasing peptide in human dental pulp. J Endod 1997; 23(3): 152-7.
14
Wurm C, Richardson JD, Bowles W, Hargreaves KM. Evaluation of functional GABA(B) receptors in dental pulp. J Endod 2001; 27(10): 620-3.
15
Mudie AS, Holland GR. Local Opioids in the Inflamed Dental Pulp. J Endod 2006; 32(4): 319-23.
16
Michaelson PL, Holland GR. Is pulpitis painful? Int Endod J 2002; 35(10): 829-32.
17
Corder G, Castro DC, Bruchas MR, Scherrer G. Endogenous and Exogenous Opioids in Pain. Annu Rev Neurosci 2018; 4: 453-73.
18
Bentley GA, Newton SH, Starr J. Evidence for an action of morphine and the enkephalins on sensory nerve endings in the mouse peritoneum. Br J Pharmacol 1981; 73(2): 325-32.
19
Schultzberg M, Lundberg JM, Hökfelt T, et al. Enkephalin-like immunoreactivity in gland cells and nerve terminals of the adrenal medulla. Neuroscience 1978; 3(12): 1169-86.
20
Joris JL, Dubner R, Hargreaves KM. Opioid analgesia at peripheral sites: a target for opioids released during stress and inflammation? Anesth Analg 1987; 66(12): 1277-81.
21
Cabot PJ, Carter L, Gaiddon C, et al. Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest 1997; 100(1): 142-8.
22
Przewłocki R, Hassan AH, Lason W, Epplen C, Herz A, Stein C. Gene expression and localization of opioid peptides in immune cells of inflamed tissue: functional role in antinociception. Neuroscience 1992; 48(2): 491-500.
23
Stein C, Hassan AH, Przewłocki R, Gramsch C, Peter K, Herz A. Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Proc Natl Acad Sci USA 1990; 87(15): 5935-9.
24
Mousa SA, Shakibaei M, Sitte N, Schäfer M, Stein C. Subcellular pathways of beta-endorphin synthesis, processing, and release from immunocytes in inflammatory pain. Endocrinology 2004; 145(3): 1331-41.
25
Basbaum A, Kaneko A, Shepherd G, Westheimer G. The Senses: A Comprehensive Reference 1st ed. 2008.
26
Stein C, Schäfer M, Machelska H. Attacking pain at its source: new perspectives on opioids. Nat Med 2003; 9(8): 1003-8.
27
Desiderio DM, Tanzer FS, Fridland G. Metabolic profiling of opioid peptides in tooth pulp by HPLC and radioreceptor assay. Neuropeptides 1985; 6(5): 463-9.
28
Robinson QC, Killmar JT, Desiderio DM, Harris EF, Fridland G. Immunoreactive evidence of beta-endorphin and methionine-enkephalin-Arg-Gly-Leu in human tooth pulp. Life Sci 1989; 45(11): 987-92.
29
Childers SR. Opioid receptor-coupled second messenger systems. Life Sci 1991; 48(21): 1991-2003.
30
Antonijevic I, Mousa SA, Schäfer M, Stein C. Perineurial defect and peripheral opioid analgesia in inflammation. J Neurosci 1995; 15(1Pt1): 165-72.
31
Barber A, Gottschlich R. Opioid agonists and antagonists: an evaluation of their peripheral actions in inflammation. Med Res Rev 1992; 12(5): 525-62.
32
Spetea M, Rydelius G, Nylander I, et al. Alteration in endogenous opioid systems due to chronic inflammatory pain conditions. Eur J Pharmacol 2002; 435(2): 245-52.
33
Mollereau C, Parmentier M, Mailleux P, et al. ORL1, a novel member of the opioid receptor family. Cloning, functional expression and localization. FEBS Lett 1994; 341(1): 33-8.
34
Gao CJ, Niu L, Ren PC, et al. Hypoxic preconditioning attenuates global cerebral ischemic injury following asphyxial cardiac arrest through regulation of delta opioid receptor system. Neuroscience 2012; 202: 352-62.
35
Husain S, Abdul Y, Potter DE. Non-analgesic effects of opioids: neuroprotection in the retina. Curr Pharm Des 2012; 18(37): 6101-8.
36
Maslov LN, Naryzhnaia NV, Tsibulnikov SY, et al. Role of endogenous opioid peptides in the infarct size-limiting effect of adaptation to chronic continuous hypoxia. Life Sci 2013; 93(9-11): 373-9.
37
Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y. Current Research on Opioid Receptor Function. Curr Drug Targets 2012; 13(2): 230-46.
38
Rusin KI, Giovannucci DR, Stuenkel EL, Moises HC. Kappa-opioid receptor activation modulates Ca2+ currents and secretion in isolated neuroendocrine nerve terminals. J Neurosci 1997; 17(17): 6565-74.
39
Zöllner C, Stein C. Opioids. Handb Exp Pharmacol 2007; (177): 31-63.
40
Fristad I, Berggreen E, Haug SR. Delta (delta) opioid receptors in small and medium-sized trigeminal neurons supporting the dental pulp of rats. Arch Oral Biol 2006; 51(4): 273-81.
41
Rechthand E, Rapoport SI. Regulation of the microenvironment of peripheral nerve: role of the blood-nerve barrier. Prog Neurobiol 1987; 28(4): 303-43.
42
Olsson Y. Microenvironment of the peripheral nervous system under normal and pathological conditions. Crit Rev Neurobiol 1990; 5(3): 265-311.
43
Herz A. Peripheral opioid analgesia-facts and mechanisms. Prog Brain Res 1996; 110: 95-104.
44
Rittner HL, Amasheh S, Moshourab R, et al. Modulation of tight junction proteins in the perineurium to facilitate peripheral opioid analgesia. Anesthesiology 2012; 116(6): 1323-34.
45
Mousa SA, Zhang Q, Sitte N, Ji R, Stein C. Beta-Endorphin-containing memory-cells and mu-opioid receptors undergo transport to peripheral inflamed tissue. J Neuroimmunol 2001; 115(1-2): 71-8.
46
Asvadi NH, Morgan M, Herath HM, Hewavitharana AK, Shaw PN, Cabot PJ. Beta-Endorphin 1–31 Biotransformation and cAMP Modulation in Inflammation. PLoS One 2014; 9(3)e90380.
47
Brack A, Rittner HL, Machelska H, et al. Endogenous peripheral antinociception in early inflammation is not limited by the number of opioid-containing leukocytes but by opioid receptor expression. Pain 2004; 108(1-2): 67-75.
48
Rittner HL, Brack A, Machelska H, et al. Opioid peptide-expressing leukocytes: identification, recruitment, and simultaneously increasing inhibition of inflammatory pain. Anesthesiology 2001; 95(2): 500-8.
49
Zollner C, Shaqura MA, Bopaiah CP, Mousa S, Stein C, Schafer M. Painful inflammation-induced increase in mu-opioid receptor binding and G-protein coupling in primary afferent neurons. Mol Pharmacol 2003; 64(2): 202-10.
50
Binder W, Mousa SA, Sitte N, Kaiser M, Stein C, Schäfer M. Sympathetic activation triggers endogenous opioid release and analgesia within peripheral inflamed tissue. Eur J Neurosci 2004; 20(1): 92-100.
51
Labuz D, Mousa SA, Schäfer M, Stein C, Machelska H. Relative contribution of peripheral versus central opioid receptors to antinociception. Brain Res 2007; 1160: 30-8.
52
Hua S, Cabot PJ. Mechanisms of peripheral immune-cell-mediated analgesia in inflammation: clinical and therapeutic implications. Trends Pharmacol Sci 2010; 31(9): 427-33.
53
Machelska H, Schopohl JK, Mousa SA, Labuz D, Schäfer M, Stein C. Different mechanisms of intrinsic pain inhibition in early and late inflammation. J Neuroimmunol 2003; 141(1-2): 30-9.
54
Stein C, Gramsch C, Herz A. Intrinsic mechanisms of antinociception in inflammation: local opioid receptors and beta-endorphin. J Neurosci 1990; 10(4): 1292-8.
55
Busch-Dienstfertig M, Stein C. Opioid receptors and opioid peptide-producing leukocytes in inflammatory pain -basic and therapeutic aspects. Brain Behav Immun 2010; 24(5): 683-94.
56
Labuz D, Berger S, Mousa SA, et al. Peripheral antinociceptive effects of exogenous and immune cell-derived endomorphins in prolonged inflammatory pain. J Neurosci 2006; 26(16): 4350-8.
57
Byers MR, Chudler EH, Ladarola MJ. Chronic tooth pulp inflammation causes transient and persistent expression of Fos in dynorphin-rich regions of rat brainstem. Brain Res 2000; 861(2): 191-207.
58
Kudo T, Chang HL, Kuroi M, Wakisaka S, Akai M, Inoki R. Influences of bradykinin and substance P on the met-enkephalin-like peptide content in the rat incisor pulp. Neuropeptides 1986; 7(4): 399-405.
59
Parris WG, Tanzer FS, Fridland GH, Harris EF, Killmar J, Desiderio DM. Effects of orthodontic force on methionine enkephalin and substance P concentrations in human pulpal tissue. Am J Orthod Dentofacial Orthop 1989; 95(6): 479-89.
60
Inoki R, Kudo T. Enkephalins and bradykinin in dental pulp. Trends Pharmacol Sci 1986; 7: 275-7.
61
Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C. Pain control in inflammation governed by selectins. Nat Med 1998; 4(12): 1425-8.
62
Sattari M, Mozayeni MA, Matloob A, Mozayeni M, Javaheri HH. Substance P and CGRP expression in dental pulps with irreversible pulpitis. Aust Endod J 2010; 36(2): 59-63.
63
Parenti C, Aricò G, Ronsisvalle G, Scoto GM. Supraspinal injection of substance P attenuates allodynia and hyperalgesia in a rat model of inflammatory pain. Peptides 2012; 34(2): 412-8.
64
Lundy FT, Linden GJ. Neuropeptides and neurogenic mechanisms in oral and periodontal inflammation. Crit Rev Oral Biol Med 2004; 15(2): 82-98.
65
Celik MÖ, Labuz D, Keye J, Glauben R, Machelska H. IL-4 induces M2 macrophages to produce sustained analgesia via opioids. JCI Insight 2020; 5(4)133093
66
Rutz JC, Hatton JF, Hildebolt C, Wells JE, Rowland KC. Localized increases in corticotropin-releasing factor receptors in pulp after dental injury. J Endod 2007; 33(11): 1319-24.
67
Smith EM, Morrill AC, Meyer WJ, Blalock JE. Corticotropin releasing factor induction of leukocyte-derived immunoreactive ACTH and endorphins. Nature 1986; 321(6073): 881-2.
68
Cabot PJ, Carter L, Schäfer M, Stein C. Methionine-enkephalin-and Dynorphin A-release from immune cells and control of inflammatory pain. Pain 2001; 93(3): 207-12.
69
Schäfer M, Carter L, Stein C. Interleukin 1 beta and corticotropin-releasing factor inhibit pain by releasing opioids from immune cells in inflamed tissue. Proc Natl Acad Sci USA 1994; 91(10): 4219-23.
70
Kavelaars A, Berkenbosch F, Croiset G, Ballieux RE, Heijnen CJ. Induction of beta-endorphin secretion by lymphocytes after subcutaneous administration of corticotropin-releasing factor. Endocrinology 1990; 126(2): 759-64.
71
Mousa SA, Bopaiah PC, Stein C, Schäfer M. Involvement of corticotropin-releasing hormone receptor subtypes 1 and 2 in peripheral opioid-mediated inhibition of inflammatory pain. Pain 2003; 106(3): 297-307.
72
Rittner HL, Labuz D, Schaefer M, et al. Pain control by CXCR2 ligands through Ca2+-regulated release of opioid peptides from polymorphonuclear cells. FASEB J 2006; 20(14): 2627-9.
73
Mousa SA, Schäfer M, Mitchell WM, Hassan AH, Stein C. Local upregulation of corticotropin-releasing hormone and interleukin-1 receptors in rats with painful hindlimb inflammation. Eur J Pharmacol 1996; 311(2-3): 221-31.
74
Gavalas A, Victoratos P, Yiangou M, Hadjipetrou-Kourounakis L, Rekka E, Kourounakis P. The anti-inflammatory effect of opioids. Int J Neurosci 1994; 74(1-4): 259-64.
75
Van der Vlist M, Raoof R, Willemen HLDM, et al. Macrophages transfer mitochondria to sensory neurons to resolve inflammatory pain. Neuron 2022; 110(4): 613-626.e9.
76
Gao L, Fan F, Wang L, et al. Polarization of macrophages in the trigeminal ganglion of rats with pulpitis. J Oral Rehabil 2022; 49(2): 228-36.
77
Da Silva MD, Bobinski F, Sato KL, Kolker SJ, Sluka KA, Santos ARS. IL-10 cytokine released from M2 macrophages is crucial for analgesic and anti-inflammatory effects of acupuncture in a model of inflammatory muscle pain. Mol Neurobiol 2015; 51(1): 19-31.
78
Vale ML, Marques JB, Moreira CA, et al. Antinociceptive effects of interleukin-4, -10, and -13 on the writhing response in mice and zymosan-induced knee joint incapacitation in rats. J Pharmacol Exp Ther 2003; 304(1): 102-8.
79
Vanderwall AG, Milligan ED. Cytokines in pain: Harnessing endogenous anti-inflammatory signaling for improved pain management. Front Immunol 2019; 10: 3009.
80
Kavelaars A, Heijnen CJ. T cells as guardians of pain resolution. Trends Mol Med 2021; 27(4): 302-13.
81
Ledeboer A, Jekich BM, Sloane EM, et al. Intrathecal interleukin-10 gene therapy attenuates paclitaxel-induced mechanical allodynia and proinflammatory cytokine expression in dorsal root ganglia in rats. Brain Behav Immun 2007; 21(5): 686-98.
82
Shen KF, Zhu HQ, Wei XH, et al. Interleukin-10 down-regulates voltage gated sodium channels in rat dorsal root ganglion neurons. Exp Neurol 2013; 247: 466-75.
83
Kimura M, Shiokawa H, Karashima Y, Sumie M, Hoka S, Yamaura K. Antinociceptive effect of selective G protein-gated inwardly rectifying K+ channel agonist ML297 in the rat spinal cord. PLoS One 2020; 15(9): e0239094.
84
Gao XF, Zhang HL, You ZD, Lu CL, He C. G protein-coupled inwardly rectifying potassium channels in dorsal root ganglion neurons. Acta Pharmacol Sin 2007; 28(2): 185-90.
85
Nockemann D, Rouault M, Labuz D, et al. The K(+) channel GIRK2 is both necessary and sufficient for peripheral opioid-mediated analgesia. EMBO Mol Med 2013; 5(8): 1263-77.
86
Karschin C, Dissmann E, Stühmer W, Karschin A. IRK(1-3) and GIRK(1-4) inwardly rectifying K+ channel mRNAs are differentially expressed in the adult rat brain. J Neurosci 1996; 16(11): 3559-70.
87
Lüscher C, Slesinger PA. Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat Rev Neurosci 2010; 11(5): 301-15.
88
Marker CL, Stoffel M, Wickman K. Spinal G-protein-gated K+ channels formed by GIRK1 and GIRK2 subunits modulate thermal nociception and contribute to morphine analgesia. J Neurosci 2004; 24(11): 2806-12.
89
Blednov YA, Stoffel M, Alva H, Harris RA. A pervasive mechanism for analgesia: activation of GIRK2 channels. Proc Natl Acad Sci USA 2003; 100(1): 277-82.
90
Nagi K, Pineyro G. Kir3 channel signaling complexes: focus on opioid receptor signaling. Front Cell Neurosci 2014; 8: 186.
91
Smith KE, Walker MW, Artymyshyn R, et al. Cloned human and rat galanin GALR3 receptors. Pharmacology and activation of G-protein inwardly rectifying K+ channels. J Biol Chem 1998; 273(36): 23321-6.
92
Dahlhaus A, Ruscheweyh R, Sandkühler J. Synaptic input of rat spinal lamina I projection and unidentified neurones in vitro. J Physiol 2005; 566(Pt2): 355-68.
93
Melnick IV. Cell type-specific postsynaptic effects of neuropeptide Y in substantia gelatinosa neurons of the rat spinal cord. Synapse 2012; 66(7): 640-9.
94
Gorham L, Just S, Doods H. Somatostatin 4 receptor activation modulates G-protein coupled inward rectifying potassium channels and voltage stimulated calcium signals in dorsal root ganglion neurons. Eur J Pharmacol 2014; 736: 101-6.
95
Engström M, Tomperi J, El-Darwish K, Ahman M, Savola JM, Wurster S. Superagonism at the human somatostatin receptor subtype 4. J Pharmacol Exp Ther 2005; 312(1): 332-8.
96
Chung MK, Cho YS, Bae YC, Lee J, Zhang X, Ro JY. Peripheral G protein-coupled inwardly rectifying potassium channels are involved in δ-opioid receptor-mediated anti-hyperalgesia in rat masseter muscle. Eur J Pain 2014; 18(1): 29-38.
97
Cichewicz DL. Synergistic interactions between cannabinoid and opioid analgesics. Life Sci 2004; 74(11): 1317-24.
98
Bushlin I, Rozenfeld R, Devi LA. Cannabinoid-opioid interactions during neuropathic pain and analgesia. Curr Opin Pharmacol 2010; 10(1): 80-6.
99
Wilson-Poe AR, Pocius E, Herschbach M, Morgan MM. The periaqueductal gray contributes to bidirectional enhancement of antinociception between morphine and cannabinoids. Pharmacol Biochem Behav 2013; 103(3): 444-9.
100
Tham SM, Angus JA, Tudor EM, Wright CE. Synergistic and additive interactions of the cannabinoid agonist CP55,940 with mu opioid receptor and alpha2-adrenoceptor agonists in acute pain models in mice. Br J Pharmacol 2005; 144(6): 875-84.
101
Roberts JD, Gennings C, Shih M. Synergistic affective analgesic interaction between delta-9-tetrahydrocannabinol and morphine. Eur J Pharmacol 2006; 530(1-2): 54-8.
102
Calignano A, La Rana G, Giuffrida A, Piomelli D. Control of pain initiation by endogenous cannabinoids. Nature 1998; 394(6690): 277-81.
103
Walker JM, Huang SM, Strangman NM, Tsou K, Sañudo-Peña MC. Pain modulation by release of the endogenous cannabinoid anandamide. Proc Natl Acad Sci USA 1999; 96(21): 12198-203.
104
Cravatt BF, Demarest K, Patricelli MP, et al. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc Natl Acad Sci USA 2001; 98(16): 9371-6.
105
Zubrzycki M, Janecka A, Liebold A, Ziegler M, Zubrzycka M. Effects of centrally administered endocannabinoids and opioids on orofacial pain perception in rats. Br J Pharmacol 2017; 174(21): 3780-9.
106
Guo J, Ikeda SR. Endocannabinoids modulate N-type calcium channels and G-protein-coupled inwardly rectifying potassium channels via CB1 cannabinoid receptors heterologously expressed in mammalian neurons. Mol Pharmacol 2004; 65(3): 665-74.
107
Klein TW, Newton C, Larsen K, et al. The cannabinoid system and immune modulation. J Leukoc Biol 2003; 74(4): 486-96.
108
Cristino L, Bisogno T, Di Marzo V. Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat Rev Neurol 2020; 16(1): 9-29.
109
Guindon J, Hohmann AG. The endocannabinoid system and pain. CNS Neurol Disord Drug Targets 2009; 8(6): 403-21.
110
Manning BH, Martin WJ, Meng ID. The rodent amygdala contributes to the production of cannabinoid-induced antinociception. Neuroscience 2003; 120(4): 1157-70.
111
Lichtman AH, Cook SA, Martin BR. Investigation of brain sites mediating cannabinoid-induced antinociception in rats: evidence supporting periaqueductal gray involvement. J Pharmacol Exp Ther 1996; 276(2): 585-93.
112
Martin WJ, Tsou K, Walker JM. Cannabinoid receptor-mediated inhibition of the rat tail-flick reflex after microinjection into the rostral ventromedial medulla. Neurosci Lett 1998; 242(1): 33-6.
113
Maccarrone M, De Petrocellis L, Bari M, et al. Lipopolysaccharide downregulates fatty acid amide hydrolase expression and increases anandamide levels in human peripheral lymphocytes. Arch Biochem Biophys 2001; 393(2): 321-8.
114
Varga K, Wagner JA, Bridgen DT, Kunos G. Platelet- and macrophage-derived endogenous cannabinoids are involved in endotoxin-induced hypotension. FASEB J 1998; 12(11): 1035-44.
115
Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990; 346(6284): 561-4.
116
Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993; 365(6441): 61-5.
117
Devane WA, Dysarz FA, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 1988; 34(5): 605-13.
118
Cravatt BF, Lichtman AH. The endogenous cannabinoid system and its role in nociceptive behavior. J Neurobiol 2004; 61(1): 149-60.
119
Howlett AC, Barth F, Bonner TI, et al. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev 2002; 54(2): 161-202.
120
Tanasescu R, Constantinescu CS. Cannabinoids and the immune system: An overview. Immunobiology 2010; 215(8): 588-97.
121
Bisogno T, Maurelli S, Melck D, De Petrocellis L, Di Marzo V. Biosynthesis, uptake, and degradation of anandamide and palmitoylethanolamide in leukocytes. J Biol Chem 1997; 272(6): 3315-23.
122
Raoof M, Sofiabadi M, Haghdoost-Yazdi H, Kooshki R, Abbasnejad M. Blockage of ventrolateral periaqueductal gray matter cannabinoid 1 receptor increases dental pulp pain and pain-related subsequent learning and memory deficits in rats. Behav Pharmacol 2022; 33(2&3): 165-74.
123
Ho YC, Lee HJ, Tung LW, et al. Activation of orexin 1 receptors in the periaqueductal gray of male rats leads to antinociception via retrograde endocannabinoid (2-Arachidonoylglycerol)-induced disinhibition. J Neurosci 2011; 31(41): 14600-10.
124
Liao HT, Lee HJ, Ho YC, Chiou LC. Capsaicin in the periaqueductal gray induces analgesia via metabotropic glutamate receptor-mediated endocannabinoid retrograde disinhibition. Br J Pharmacol 2011; 163(2): 330-45.
125
Behbehani MM, Jiang M, Chandler SD, Ennis M. The effect of GABA and its antagonists on midbrain periaqueductal gray neurons in the rat. Pain 1990; 40(2): 195-204.
126
Felder CC, Joyce KE, Briley EM, et al. Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Mol Pharmacol 1995; 48(3): 443-50.
127
Ibsen MS, Connor M, Glass M. Cannabinoid CB1 and CB2 receptor signaling and bias. Cannabis Cannabinoid Res 2017; 2(1): 48-60.
128
Ross HR, Gilmore AJ, Connor M. Inhibition of human recombinant T-type calcium channels by the endocannabinoid N-arachidonoyl dopamine. Br J Pharmacol 2009; 156(5): 740-50.
129
O’Sullivan SE, Kendall DA. Cannabinoid activation of peroxisome proliferator-activated receptors: potential for modulation of inflammatory disease. Immunobiology 2010; 215(8): 611-6.
130
Miyashita K, Oyama T, Sakuta T, Tokuda M, Torii M. Anandamide induces matrix metalloproteinase-2 production through cannabinoid-1 receptor and transient receptor potential vanilloid-1 in human dental pulp cells in culture. J Endod 2012; 38(6): 786-90.
131
Kissin I, Szallasi A. Therapeutic targeting of TRPV1 by resiniferatoxin, from preclinical studies to clinical trials. Curr Top Med Chem 2011; 11(17): 2159-70.
132
Engel MA, Izydorczyk I, Mueller-Tribbensee SM, Becker C, Neurath MF, Reeh PW. Inhibitory CB1 and activating/desensitizing TRPV1-mediated cannabinoid actions on CGRP release in rodent skin. Neuropeptides 2011; 45(3): 229-37.
133
Maione S, Bisogno T, De Novellis V, et al. Elevation of endocannabinoid levels in the ventrolateral periaqueductal grey through inhibition of fatty acid amide hydrolase affects descending nociceptive pathways via both cannabinoid receptor type 1 and transient receptor potential vanilloid type-1 receptors. J Pharmacol Exp Ther 2006; 316(3): 969-82.
134
Palazzo E, Luongo L, De Novellis V, Berrino L, Rossi F, Maione S. Moving towards supraspinal TRPV1 receptors for chronic pain relief. Mol Pain 2010; 6: 66.
135
McCarberg BH, Barkin RL. The future of cannabinoids as analgesic agents: a pharmacologic, pharmacokinetic, and pharmacodynamic overview. Am J Ther 2007; 14(5): 475-83.
136
Mbvundula EC, Rainsford KD, Bunning RAD. Cannabinoids in pain and inflammation. Inflammopharmacology 2004; 12(2): 99-114.
137
Akopian AN, Ruparel NB, Patwardhan A, Hargreaves KM. Cannabinoids Desensitize Capsaicin and Mustard Oil Responses in Sensory Neurons via TRPA1 Activation. J Neurosci 2008; 28(5): 1064-75.
138
Price TJ, Patwardhan A, Akopian AN, Hargreaves KM, Flores CM. Modulation of trigeminal sensory neuron activity by the dual cannabinoid-vanilloid agonists anandamide, N-arachidonoyl-dopamine and arachidonyl-2-chloroethylamide. Br J Pharmacol 2004; 141(7): 1118-11130.
139
Jordt SE, Bautista DM, Chuang HH, et al. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 2004; 427(6971): 260-5.
140
Liu L, Lo Y, Chen I, Simon SA. The responses of rat trigeminal ganglion neurons to capsaicin and two nonpungent vanilloid receptor agonists, olvanil and glyceryl nonamide. J Neurosci 1997; 17(11): 4101-11.
141
Tsumura M, Sobhan U, Muramatsu T, et al. TRPV1-mediated calcium signal couples with cannabinoid receptors and sodium-calcium exchangers in rat odontoblasts. Cell Calcium 2012; 52(2): 124-36.
142
Que K, He D, Jin Y, et al. Expression of Cannabinoid Type 1 Receptors in Human Odontoblast Cells. J Endod 2017; 43(2): 283-8.
143
Nikolaeva EP, Cox TC, Flake NM. Osseous characteristics of mice lacking cannabinoid receptor 2 after pulp exposure. J Endod 2015; 41(6): 853-7.
144
McPartland JM. Expression of the endocannabinoid system in fibroblasts and myofascial tissues. J Bodyw Mov Ther 2008; 12(2): 169-82.
145
Qi X, Liu C, Li G, et al. Evaluation of cannabinoids on the odonto/osteogenesis in human dental pulp cells in vitro. J Endod 2021; 47(3): 444-50.
146
Matias I, Gonthier MP, Orlando P, et al. Regulation, function, and dysregulation of endocannabinoids in models of adipose and beta-pancreatic cells and in obesity and hyperglycemia. J Clin Endocrinol Metab 2006; 91(8): 3171-80.
147
Richardson D, Pearson RG, Kurian N, et al. Characterisation of the cannabinoid receptor system in synovial tissue and fluid in patients with osteoarthritis and rheumatoid arthritis. Arthritis Res Ther 2008; 10(2): R43.
148
McPartland JM, Skinner E. The biodynamic model of osteopathy in the cranial field. Explore (NY) 2005; 1(1): 21-32.
149
Flake NM, Zweifel LS. Behavioral Effects of Pulp Exposure in Mice Lacking Cannabinoid Receptor 2. J Endod 2012; 38(1): 86-90.
150
Guindon J, Hohmann AG. Cannabinoid CB2 receptors: a therapeutic target for the treatment of inflammatory and neuropathic pain. Br J Pharmacol 2008; 153(2): 319-34.
151
Turcotte C, Blanchet MR, Laviolette M, Flamand N. The CB2 receptor and its role as a regulator of inflammation. Cell Mol Life Sci 2016; 73(23): 4449-70.
152
Tang Y, Wolk B, Britch SC, Craft RM, Kendall DA. Anti-inflammatory and antinociceptive effects of the selective cannabinoid CB2 receptor agonist ABK5. J Pharmacol Sci 2021; 145(4): 319-26.
153
Correa F, Mestre L, Docagne F, Guaza C. Activation of cannabinoid CB2 receptor negatively regulates IL-12p40 production in murine macrophages: role of IL-10 and ERK1/2 kinase signaling. Br J Pharmacol 2005; 145(4): 441-8.
154
Yuan M, Kiertscher SM, Cheng Q, Zoumalan R, Tashkin DP, Roth MD. Delta 9-Tetrahydrocannabinol regulates Th1/Th2 cytokine balance in activated human T cells. J Neuroimmunol 2002; 133(1-2): 124-31.
155
Börner C, Bedini A, Höllt V, Kraus J. Analysis of promoter regions regulating basal and interleukin-4-inducible expression of the human CB1 receptor gene in T lymphocytes. Mol Pharmacol 2008; 73(3): 1013-9.
156
Jean-Gilles L, Gran B, Constantinescu CS. Interaction between cytokines, cannabinoids and the nervous system. Immunobiology 2010; 215(8): 606-10.
157
Crowley T, Cryan JF, Downer EJ, O’Leary OF. Inhibiting neuroinflammation: The role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun 2016; 54: 260-77.
158
Beltrán González AN, López Pazos MI, Calvo DJ. Reactive oxygen species in the regulation of the GABA mediated inhibitory neurotransmission. Neuroscience 2020; 439: 137-45.
159
Goudet C, Magnaghi V, Landry M, Nagy F, Gereau R, Pin J. Metabotropic receptors for glutamate and GABA in pain. Brain Res Brain Res Rev 2009; 60(1): 43-56.
160
Parker DAS, Marino V. GABA heteroreceptors modulate noradrenaline release in human dental pulp. J Dent Res 2013; 92(11): 1017-21.
161
Wu LA, Huang J, Wang W, et al. Activation of GABAergic neurons following tooth pulp stimulation. J Dent Res 2010; 89(5): 532-6.
162
Takeda M, Tanimoto T, Ikeda M, Kadoi J, Matsumoto S. Activation of GABAB receptor inhibits the excitability of rat small diameter trigeminal root ganglion neurons. Neuroscience 2004; 123(2): 491-505.
163
Ranjbar Ekbatan M, Cairns BE. Attenuation of sensory transmission through the rat trigeminal ganglion by GABA receptor activation. Neuroscience 2021; 471: 80-92.
164
Nassery K, Marino V, Parker DAS. Uptake and release of [3H]GABA in human dental pulp. Arch Oral Biol 2007; 52(7): 607-13.
165
Nowak P, Kowalińska-Kania M, Nowak D, Kostrzewa RM, Malinowska-Borowska J. Antinociceptive effects of H3 (R-methylhistamine) and GABA(B) (baclofen)-receptor ligands in an orofacial model of pain in rats. Neurotox Res 2013; 24(2): 258-64.
166
Whitehead RA, Puil E, Ries CR, et al. GABA(B) receptor-mediated selective peripheral analgesia by the non-proteinogenic amino acid, isovaline. Neuroscience 2012; 213: 154-60.
167
Oshima K, Takeda M, Tanimoto T, Katsuumi I, Matsumoto S. Tooth-pulp-evoked rostral spinal trigeminal neuronal excitation is attenuated by the activation of 5-HT3 receptors via GABAergic interneurons in the rat. Brain Res 2006; 1109(1): 70-3.
168
Oshima K, Takeda M, Tanimoto T, Katsuumi I, Matsumoto S. Tooth-pulp-evoked rostral spinal trigeminal nucleus neuron activity is inhibited by conditioning sciatic nerve stimulation in the rat: possible role of 5-HT3 receptor mediated GABAergic inhibition. Brain Res Bull 2005; 65(1): 31-40.
169
Caviedes-Bucheli J, Muñoz HR, Azuero-Holguín MM, Ulate E. Neuropeptides in dental pulp: the silent protagonists. J Endod 2008; 34(7): 773-88.
170
Abrams GM, Recht L. Neuropeptides and their role in pain and analgesia. Acupunct Electrother Res 1982; 7(2-3): 105-21.
171
Gamse R, Leeman SE, Holzer P, Lembeck F. Differential effects of capsaicin on the content of somatostatin, substance P, and neurotensin in the nervous system of the rat. Naunyn Schmiedebergs Arch Pharmacol 1981; 317(2): 140-8.
172
Patel YC. Somatostatin and its receptor family. Front Neuroendocrinol 1999; 20(3): 157-98.
173
Schuelert N, Just S, Kuelzer R, Corradini L, Gorham LCJ, Doods H. The somatostatin receptor 4 agonist J-2156 reduces mechanosensitivity of peripheral nerve afferents and spinal neurons in an inflammatory pain model. Eur J Pharmacol 2015; 746: 274-81.
174
Hökfelt T, Elde R, Johansson O, Luft R, Nilsson G, Arimura A. Immunohistochemical evidence for separate populations of somatostatin-containing and substance P-containing primary afferent neurons in the rat. Neuroscience 1976; 1(2): 131-6.
175
Pintér E, Helyes Z, Szolcsányi J. Inhibitory effect of somatostatin on inflammation and nociception. Pharmacol Ther 2006; 112(2): 440-56.
176
Patel YC, Greenwood MT, Panetta R, Demchyshyn L, Niznik H, Srikant CB. The somatostatin receptor family. Life Sci 1995; 57(13): 1249-65.
177
Song YH, Yoon J, Lee SH. The role of neuropeptide somatostatin in the brain and its application in treating neurological disorders. Exp Mol Med 2021; 53(3): 328-38.
178
Hoyer D, Pérez J, Schoeffter P, et al. Pharmacological identity between somatostatin SS-2 binding sites and SSTR-1 receptors. Eur J Pharmacol 1995; 289(1): 151-61.
179
Green PG, Basbaum AI, Levine JD. Sensory neuropeptide interactions in the production of plasma extravasation in the rat. Neuroscience 1992; 50(3): 745-9.
180
Morton CR, Hutchison WD, Hendry IA, Duggan AW. Somatostatin: evidence for a role in thermal nociception. Brain Res 1989; 488(1-2): 89-96.
181
Szolcsányi J, Bölcskei K, Szabó A, et al. Analgesic effect of TT-232, a heptapeptide somatostatin analogue, in acute pain models of the rat and the mouse and in streptozotocin-induced diabetic mechanical allodynia. Eur J Pharmacol 2004; 498(1-3): 103-9.
182
Szolcsányi J, Pintér E, Helyes Z, Oroszi G, Németh J. Systemic anti-inflammatory effect induced by counter-irritation through a local release of somatostatin from nociceptors. Br J Pharmacol 1998; 125(4): 916-22.
183
Jian K, Barhoumi R, Ko ML, Ko GY. Inhibitory effect of somatostatin-14 on L-type voltage-gated calcium channels in cultured cone photoreceptors requires intracellular calcium. J Neurophysiol 2009; 102(3): 1801-10.
184
Mihara S, North RA, Surprenant A. Somatostatin increases an inwardly rectifying potassium conductance in guinea-pig submucous plexus neurones. J Physiol 1987; 390: 335-55.
185
Moore SD, Madamba SG, Joëls M, Siggins GR. Somatostatin augments the M-current in hippocampal neurons. Science 1988; 239(4837): 278-80.
186
Wang HL, Bogen C, Reisine T, Dichter M. Somatostatin-14 and somatostatin-28 induce opposite effects on potassium currents in rat neocortical neurons. Proc Natl Acad Sci USA 1989; 86(23): 9616-20.
187
Toossi H, Del Cid-Pellitero E, Stroh T, Jones BE. Somatostatin varicosities contain the vesicular GABA transporter and contact orexin neurons in the hypothalamus. Eur J Neurosci 2012; 36(10): 3388-95.
188
Randić M, Miletić V. Depressant actions of methionine-enkephalin and somatostatin in cat dorsal horn neurones activated by noxious stimuli. Brain Res 1978; 152(1): 196-202.
189
Sicuteri F, Rainò L, Geppetti P. Substance P and endogenous opioids: how and where they could play a role in cluster headache. Cephalalgia Int J Headache 1983; 3(Suppl. 1): 143-5.
190
Scicchitano R, Dazin P, Bienenstock J, Payan DG, Stanisz AM. The murine IgA-secreting plasmacytoma MOPC-315 expresses somatostatin receptors. J Immunol 1988; 141(3): 937-41.
191
Casasco A, Calligaro A, Casasco M, Springall DR, Polak JM, Marchetti C. Immunocytochemical Evidence for the Presence of Somatostatin-like Immunoreactive Nerves in Human Dental Pulp. J Dent Res 1991; 70(2): 87-9.
192
Selmer I, Schindler M, Allen JP, Humphrey PP, Emson PC. Advances in understanding neuronal somatostatin receptors. Regul Pept 2000; 90(1-3): 1-18.
193
ten Bokum AM, Hofland LJ, van Hagen PM. Somatostatin and somatostatin receptors in the immune system: a review. Eur Cytokine Netw 2000; 11(2): 161-76.
194
Lawson SN. Neuropeptides in morphologically and functionally identified primary afferent neurons in dorsal root ganglia: substance P, CGRP and somatostatin. Prog Brain Res 1995; 104: 161-73.
195
Gonzalez-Rey E, Varela N, Sheibanie AF, Chorny A, Ganea D, Delgado M. Cortistatin, an antiinflammatory peptide with therapeutic action in inflammatory bowel disease. Proc Natl Acad Sci USA 2006; 103(11): 4228-33.
196
Spier AD, de Lecea L. Cortistatin: a member of the somatostatin neuropeptide family with distinct physiological functions. Brain Res Brain Res Rev 2000; 33(2-3): 228-41.
197
Morell M, Camprubí-Robles M, Culler MD, de Lecea L, Delgado M. Cortistatin attenuates inflammatory pain via spinal and peripheral actions. Neurobiol Dis 2014; 63: 141-54.
198
Robas N, Mead E, Fidock M. MrgX2 is a high potency cortistatin receptor expressed in dorsal root ganglion. J Biol Chem 2003; 278(45): 44400-4.
199
Flood JF, Uezu K, Morley JE. The cortical neuropeptide, cortistatin-14, impairs post-training memory processing. Brain Res 1997; 775(1-2): 250-2.
200
De Lecea L, del Rio JA, Criado JR, et al. Cortistatin is expressed in a distinct subset of cortical interneurons. J Neurosci 1997; 17(15): 5868-80.
201
Dalm VA, Van Hagen PM, Van Koetsveld PM, et al. Cortistatin rather than somatostatin as a potential endogenous ligand for somatostatin receptors in the human immune system. J Clin Endocrinol Metab 2003; 88(1): 270-6.
202
Dalm VASH, Van Hagen PM, Van Koetsveld PM, Achilefu S, Houtsmuller AB, Pols DHJ. van der, Lely A.; J. Lamberts, S.; Hofland, LJ. Expression of somatostatin, cortistatin, and somatostatin receptors in human monocytes, macrophages, and dendritic cells. Am J Physiol Endocrinol Metab 2003; 285(2): 344-53.
203
Duran-Prado M, Morell M, Delgado-Maroto V, et al. Culler.; Hernandez-Cortes P.; O’Valle F.; Delgado M. Cortistatin inhibits migration and proliferation of human vascular smooth muscle cells and decreases neointimal formation on carotid artery ligation. Circ Res 2013; 112(11): 1444-55.
204
Gonzalez-Rey E, Delgado M. Cortistatin as a potential multistep therapeutic agent for inflammatory disorders. Drug News Perspect 2006; 19(7): 393-9.
205
Capuano A, Currò D, Navarra P, Tringali G. Cortistatin modulates calcitonin gene-related peptide release from neuronal tissues of rat. Comparison with somatostatin. Peptides 2011; 32(1): 138-43.
206
De Lecea L, Criado JR, Prospero-Garcia O, et al. A cortical neuropeptide with neuronal depressant and sleep-modulating properties. Nature 1996; 381(6579): 242-5.
207
Liu Y, Zhou YB, Zhang GG, et al. Cortistatin attenuates vascular calcification in rats. Regul Pept 2010; 159(1-3): 35-43.
208
Delgado M, Gonzalez-Rey E. Role of Cortistatin in the Stressed Immune System 2017.
209
Gonzalez-Rey E, Chorny A, Delgado M. Regulation of immune tolerance by anti-inflammatory neuropeptides. Nat Rev Immunol 2007; 7(1): 52-63.
210
Falo CP, Benitez R, Caro M, et al. The Neuropeptide Cortistatin Alleviates Neuropathic Pain in Experimental Models of Peripheral Nerve Injury. Pharmaceutics 2021; 13(7): 947.
211
Souza-Moreira L, Morell M, Delgado-Maroto V, et al. Paradoxical effect of cortistatin treatment and its deficiency on experimental autoimmune encephalomyelitis. J Immunol 2013; 191(5): 2144-54.
212
Pääkkönen V, Bleicher F, Carrouel F, et al. General expression profiles of human native odontoblasts and pulp-derived cultured odontoblast-like cells are similar but reveal differential neuropeptide expression levels. Arch Oral Biol 2009; 54(1): 55-62.
213
Ichikawa H, Helke CJ. Distribution, origin and plasticity of galanin-immunoreactivity in the rat carotid body. Neuroscience 1993; 52(3): 757-67.
214
Melander T, Hökfelt T, Rökaeus A. Distribution of galaninlike immunoreactivity in the rat central nervous system. J Comp Neurol 1986; 248(4): 475-517.
215
Skofitsch G, Jacobowitz DM. Galanin-like immunoreactivity in capsaicin sensitive sensory neurons and ganglia. Brain Res Bull 1985; 15(2): 191-5.
216
Lang R, Gundlach AL, Holmes FE, et al. Physiology, Signaling, and Pharmacology of Galanin Peptides and Receptors: Three Decades of Emerging Diversity. Pharmacol Rev 2015; 67(1): 118-75.
217
Bauer JW, Lang R, Jakab M, Kofler B. Galanin family of peptides in skin function. Exp Suppl 2012; 2010(102): 51-9.
218
Lang R, Kofler B. The galanin peptide family in inflammation. Neuropeptides 2011; 45(1): 1-8.
219
Hobson S-A, Holmes FE, Kerr NCH, Pope RJP, Wynick D. Mice deficient for galanin receptor 2 have decreased neurite outgrowth from adult sensory neurons and impaired pain-like behaviour. J Neurochem 2006; 99(3): 1000-10.
220
Branchek TA, Smith KE, Gerald C, Walker MW. Galanin receptor subtypes. Trends Pharmacol Sci 2000; 21(3): 109-17.
221
Habert-Ortoli E, Amiranoff B, Loquet I, Laburthe M, Mayaux JF. Molecular cloning of a functional human galanin receptor. Proc Natl Acad Sci USA 1994; 91(21): 9780-3.
222
Howard AD, Tan C, Shiao LL, et al. Molecular cloning and characterization of a new receptor for galanin. FEBS Lett 1997; 405(3): 285-90.
223
Wang S, Hashemi T, Fried S, Clemmons AL, Hawes BE. Differential intracellular signaling of the GalR1 and GalR2 galanin receptor subtypes. Biochemistry 1998; 37(19): 6711-7.
224
Lang R, Gundlach AL, Kofler B. The galanin peptide family: receptor pharmacology, pleiotropic biological actions, and implications in health and disease. Pharmacol Ther 2007; 115(2): 177-207.
225
Yu L-C, Xu S-L, Xiong W, Lundeberg T. The effect of galanin on wide-dynamic range neuron activity in the spinal dorsal horn of rats. Regul Pept 2001; 101(1): 179-82.
226
Liu H-X, Hökfelt T. The participation of galanin in pain processing at the spinal level. Trends Pharmacol Sci 2002; 23(10): 468-74.
227
Hua X-Y, Hayes CS, Hofer A, et al. Galanin acts at GalR1 receptors in spinal antinociception: synergy with morphine and AP-5. J Pharmacol Exp Ther 2004; 308(2): 574-82.
228
Wiesenfeld-Hallin Z, Xu X-J, Crawley JN, Hökfelt T. Galanin and spinal nociceptive mechanisms: Recent results from transgenic and knock-out models. Neuropeptides 2005; 39(3): 207-10.
229
Xu X-J, Hökfelt T, Wiesenfeld-Hallin Z. Galanin – 25 years with a multitalented neuropeptide. Cell Mol Life Sci 2008; 65(12): 1813-9.
230
Jin W-Y, Liu Z, Liu D, Yu L-C. Antinociceptive effects of galanin in the central nucleus of amygdala of rats, an involvement of opioid receptors. Brain Res 2010; 1320: 16-21.
231
Endoh T, Sato D, Wada Y, et al. Galanin inhibits calcium channels via Gαi-protein mediated by GalR1 in rat nucleus tractus solitarius. Brain Res 2008; 1229: 37-46.
232
Hao JX, Shi TJ, Xu IS, et al. Intrathecal galanin alleviates allodynia-like behaviour in rats after partial peripheral nerve injury. Eur J Neurosci 1999; 11(2): 427-32.
233
Yang Y, Zhang Y, Li XH, et al. Involvements of galanin and its receptors in antinociception in nucleus accumbens of rats with inflammatory pain. Neurosci Res 2015; 97: 20-5.
234
Reimann W, Englberger W, Friderichs E, Selve N, Wilffert B. Spinal antinociception by morphine in rats is antagonized by galanin receptor antagonists. Naunyn Schmiedebergs Arch Pharmacol 1994; 350(4): 380-6.
235
Wiesenfeld-Hallin Z, Xu XJ, Langel U, Bedecs K, Hökfelt T, Bartfai T. Galanin-mediated control of pain: enhanced role after nerve injury. Proc Natl Acad Sci USA 1992; 89(8): 3334-7.
236
Brumovsky P, Mennicken F, O’donnell D, Hökfelt T. Differential distribution and regulation of galanin receptors- 1 and -2 in the rat lumbar spinal cord. Brain Res 2006; 1085(1): 111-20.
237
Landry M, Bouali-Benazzouz R, André C, et al. Galanin receptor 1 is expressed in a subpopulation of glutamatergic interneurons in the dorsal horn of the rat spinal cord. J Comp Neurol 2006; 499(3): 391-403.
238
Yue H-Y, Fujita T, Kumamoto E. Biphasic modulation by galanin of excitatory synaptic transmission in substantia gelatinosa neurons of adult rat spinal cord slices. J Neurophysiol 2011; 105(5): 2337-49.
239
Wittau N, Grosse R, Kalkbrenner F, Gohla A, Schultz G, Gudermann T. The galanin receptor type 2 initiates multiple signaling pathways in small cell lung cancer cells by coupling to G(q), G(i) and G(12) proteins. Oncogene 2000; 19(37): 4199-209.
240
Xiong W, Gao L, Sapra A, Yu L-C. Antinociceptive role of galanin in the spinal cord of rats with inflammation, an involvement of opioid systems. Regul Pept 2005; 132(1): 85-90.
241
Suzuki H, Iwanaga T, Yoshie H, et al. Expression of galanin receptor-1 (GALR1) in the rat trigeminal ganglia and molar teeth. Neurosci Res 2002; 42(3): 197-207.
242
Byers MR. Dental sensory receptors. Int Rev Neurobiol 1984; 25: 39-94.
243
Kerr BJ, Gupta Y, Pope R, Thompson SWN, Wynick D, McMahon SB. Endogenous galanin potentiates spinal nociceptive processing following inflammation. Pain 2001; 93(3): 267-77.
244
Reeve AJ, Walker K, Urban L, Fox A. Excitatory effects of galanin in the spinal cord of intact, anaesthetized rats. Neurosci Lett 2000; 295(1-2): 25-8.
245
Liu F, Yajima T, Wang M, Shen J-F, Ichikawa H, Sato T. Effects of trigeminal nerve injury on the expression of galanin and its receptors in the rat trigeminal ganglion. Neuropeptides 2020; 84102098
246
Liu HX, Brumovsky P, Schmidt R, et al. Dios mío, C.; Hökfelt, T. Receptor subtype-specific pronociceptive and analgesic actions of galanin in the spinal cord: selective actions via GalR1 and GalR2 receptors. Proc Natl Acad Sci USA 2001; 98(17): 9960-4.
247
Jimenez-Andrade JM, Zhou S, Yamaxni A, Valencia de Ita S, Castañeda-Hernandez G, Carlton SM. Mechanism by which peripheral galanin increases acute inflammatory pain. Brain Res 2005; 1056(2): 113-7.
248
Hulse RP, Donaldson LF, Wynick D. Peripheral galanin receptor 2 as a target for the modulation of pain. Pain Res Treat 2012; 2012545386
249
Pert A, Moody TW, Pert CB, Dewald LA, Rivier J. Bombesin: receptor distribution in brain and effects on nociception and locomotor activity. Brain Res 1980; 193(1): 209-20.
250
Zhang HP, Xiao Z, Cilz NI, Hu B, Dong H, Lei S. Bombesin facilitates GABAergic transmission and depresses epileptiform activity in the entorhinal cortex. Hippocampus 2014; 24(1): 21-31.
251
Jensen RT, Battey JF, Spindel ER, Benya RV. International Union of Pharmacology. LXVIII. Mammalian bombesin receptors: nomenclature, distribution, pharmacology, signaling, and functions in normal and disease states. Pharmacol Rev 2008; 60(1): 1-42.
252
King B, Jones M, Ewart W. Immunocytochemical localization of bombesin-like peptides in afferent cranial nerves and brain stem nuclei in rats. Ann N Y Acad Sci 1988; 547: 447-7.
253
Aubeux D, Peters O, Hosseinpour S, et al. Specialized pro-resolving lipid mediators in endodontics: a narrative review. BMC Oral Health 2021; 21(1): 276.
254
Cotti E, Ideo F, Pedrazzini A, Bardini G, Musu D, Kantarci A. Proresolving mediators in endodontics: A systematic review. J Endod 2021; 47(5): 711-20.
255
Dalli J. Does promoting resolution instead of inhibiting inflammation represent the new paradigm in treating infections? Mol Aspects Med 2017; 58: 12-20.
256
Chiang N, Serhan CN. Structural elucidation and physiologic functions of specialized pro-resolving mediators and their receptors. Mol Aspects Med 2017; 58: 114-29.
257
Fattori V, Zaninelli TH, Rasquel-Oliveira FS, Casagrande R, Verri WA. Specialized pro-resolving lipid mediators: A new class of non-immunosuppressive and non-opioid analgesic drugs. Pharmacol Res 2020; 151104549
258
Ji RR, Xu ZZ, Strichartz G, Serhan CN. Emerging Roles of Resolvins in the Resolution of Inflammation and Pain. Trends Neurosci 2011; 34(11): 599-609.
259
Sommer C, Birklein F. Fighting off pain with resolvins. Nat Med 2010; 16(5): 518-20.
260
Yoo S, Lim JY, Hwang SW. Resolvins: Endogenously-generated potent painkilling substances and their therapeutic perspectives. Curr Neuropharmacol 2013; 11(6): 664-76.
261
Roh J, Go EJ, Park JW, Kim YH, Park C. Resolvins: Potent Pain Inhibiting Lipid Mediators via Transient Receptor Potential Regulation. Front Cell Dev Biol 2020; 8584206
262
Xu ZZ, Zhang L, Liu T, et al. Resolvins RvE1 and RvD1Attenuate Inflammatory Pain via Central and Peripheral Actions. Nat Med 2010; 16(5): 592-7.
263
Leuti A, Fava M, Pellegrini N, Maccarrone M. Role of specialized pro-resolving mediators in neuropathic pain. Front Pharmacol 2021; 12717993
264
Zhang LY, Jia MR, Sun T. The roles of special proresolving mediators in pain relief. Rev Neurosci 2018; 29(6): 645-60.
265
Tao X, Lee MS, Donnelly CR, Ji R. Neuromodulation, Specialized Proresolving Mediators, and Resolution of Pain. Neurotherapeutics 2020; 17(3): 886-99.
266
Fredman G, Serhan C. Specialized pro‐resolving mediators: wiring the circuitry of effector immune and tissue homeostasis. Endod Topics 2011; 24(1): 39-58.
267
Serhan CN, Chiang N. Resolution phase lipid mediators of inflammation: agonists of resolution. Curr Opin Pharmacol 2013; 13(4): 32-40.
268
Keinan D, Leigh NJ, Nelson JW, de Oleo L, Baker O. Understanding resolvin signaling pathways to improve oral health. Int J Mol Sci 2013; 14(3): 5501-18.
269
Sommer C, Birklein F. Resolvins and inflammatory pain. F1000 Med Rep 2011; 3: 19.
270
Khan J, Puchimada B, Kadouri D, Zusman T, Javed F, Eliav E. The anti-nociceptive effects of Porphyromonas gingivalis lipopolysaccharide. Arch Oral Biol 2019; 102: 193-8.
271
Deng L, Chiu I. Microbes and pain. PLoS Pathog 2021; 17e1009398
272
Ko Yj, Ky K, Ky K, et al. The Anti-Inflammatory Effect of Human Telomerase-Derived Peptide on P. gingivalis Lipopolysaccharide-Induced Inflammatory Cytokine Production and Its Mechanism in Human Dental Pulp Cells. Mediators Inflamm 2015; 2015: 385127.
273
Martin FE, Nadkarni MA, Jacques NA, Hunter N. Quantitative microbiological study of human carious dentine by culture and real-time PCR: association of anaerobes with histopathological changes in chronic pulpitis. J Clin Microbiol 2002; 40(5): 1698-704.
274
Sundqvist G, Johansson E, Sjögren U. Prevalence of black-pigmented bacteroides species in root canal infections. J Endod 1989; 15(1): 13-9.
275
Milshteyn A, Colosimo DA, Brady SF. Accessing bioactive natural products from the human microbiome. Cell Host Microbe 2018; 23(6): 725-36.
276
Structure, function and diversity of the healthy human microbiome. Nature 2012; 486(7402): 207-14.
277
Qin J. Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.; Manichanh, C.; Nielsen, T.; Pons, N.; Levénez, F.; Yamada, T.; Mende, D.; Li, J.; Xu, J.; Li, S.; Li, D.; Cao, J.; Wang, B.; Liang, H.; Zheng, H.; Xie, Y.; Lepage, P.; Bertalán, M.; Batto, J.; Hansen, J.; Le Paslier, D.; Linneberg, A.; Nielsen, HB.; Pelletier, E.; Renault, P.; Sicheritz-Ponten, T.; Turner, K.; Zhu, H.; Yu, C.; Li, S.; Zhou, Y.; Li, Y.; Xiuqing, Z.; Li, S.; Yang, H.; Wang, J.; Brunak, S.; Guarner, F.; Kristiansen, K.; Pedersen, O.; Weissenbach, J.; MetaHIT, C.; Bork, P.; Ehrlich, D.; Wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010; 464(7285): 59-65.
278
O’Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology 2005; 128(3): 541-51.
279
Verdú EF, Bercik P, Verma-Gandhu M, et al. Rochat.; Collins, SM. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut 2006; 55(2): 182-90.
280
Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med 2007; 13(1): 35-7.
281
Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J Appl Microbiol 2012; 113(2): 411-7.
282
Duranti S, Ruiz L, Lugli GA, et al. Bifidobacterium adolescentis as a key member of the human gut microbiota in the production of GABA. Sci Rep 2020; 10(1): 14112.
283
Pokusaeva K, Johnson C, Luk B, et al. lugo.; La mayor.; Mori-Akiyama Y.; Hollister EB.; dan sm.; Shi XZ.; Engler DA.; Savidge T.; Versalovich J. GABA-producing Bifidobacterium dentium modulates visceral sensitivity in the intestine. Neurogastroenterol Motil Off J Eur Gastrointest Motil Soc 2017; 29(1): e12904.
284
Donnelly CR, Chen O, Ji RR. How Do Sensory Neurons Sense Danger Signals? Trends Neurosci 2020; 43(10): 822-38.
285
Chiu IM, Heesters BA, Ghasemlou N, et al. Wardenburg.; Hwang, S.; Carroll, M.; Woolf, C. Bacteria activate sensory neurons that modulate pain and inflammation. Nature 2013; 501(7465): 52-7.
286
Diogenes A, Ferraz CCR, Akopian AN, Henry MA, Hargreaves KM. LPS sensitizes TRPV1 via activation of TLR4 in trigeminal sensory neurons. J Dent Res 2011; 90(6): 759-64.
287
Gong L, Gao F, Li J, et al. Oxytocin-induced membrane hyperpolarization in pain-sensitive dorsal root ganglia neurons mediated by Ca(2+)/nNOS/NO/KATP pathway. Neuroscience 2015; 289: 417-28.
288
Yu SQ, Lundeberg T, Yu LC. Involvement of oxytocin in spinal antinociception in rats with inflammation. Brain Res 2003; 983(1-2): 13-22.
289
Ando M, Hayashi Y, Hitomi S, et al. Oxytocin-Dependent Regulation of TRPs Expression in Trigeminal Ganglion Neurons Attenuates Orofacial Neuropathic Pain following Infraorbital Nerve Injury in Rats. Int J Mol Sci 2020; 21(23): 9173.
290
Warfvinge K, Krause DN, Maddahi A, et al. Oxytocin as a regulatory neuropeptide in the trigeminovascular system: Localization, expression and function of oxytocin and oxytocin receptors. Cephalalgia 2020; 40(12): 1283-95.
291
Tzabazis A, Mechanic J, Miller J, et al. Oxytocin receptor: Expression in the trigeminal nociceptive system and potential role in the treatment of headache disorders. Cephalalgia 2016; 36(10): 943-50.
292
Schorscher-Petcu A, Sotocinal S, Ciura S, et al. Oxytocin-Induced Analgesia and Scratching Are Mediated by the Vasopressin-1A Receptor in the Mouse. J Neurosci 2010; 30(24): 8274-84.
293
Qiu F, Qiu CY, Cai H, et al. Oxytocin inhibits the activity of acid-sensing ion channels through the vasopressin, V1A receptor in primary sensory neurons. Br J Pharmacol 2014; 171(12): 3065-76.
294
Manzano-García A, González-Hernández A, Tello-García IA, Martínez-Lorenzana G, Condés-Lara M. The role of peripheral vasopressin 1A and oxytocin receptors on the subcutaneous vasopressin antinociceptive effects. Eur J Pain 2018; 22(3): 511-26.
295
Kubo A, Shinoda M, Katagiri A, et al. Oxytocin alleviates orofacial mechanical hypersensitivity associated with infraorbital nerve injury through vasopressin-1A receptors of the rat trigeminal ganglia. Pain 2017; 158(4): 649-59.
296
Qiu F, Hu WP, Yang ZF. Enhancement of GABA-activated currents by arginine vasopressin in rat dorsal root ganglion neurons. Sheng Li Xue Bao 2014; 66(6): 647-57.
297
Nersesyan Y, Demirkhanyan L, Cabezas-Bratesco D, et al. Oxytocin modulates nociception as an agonist of pain-sensing TRPV1. Cell Rep 2017; 21(6): 1681-91.
298
Lyu RM, Huang XF, Zhang Y, et al. Phoenixin: a novel peptide in rodent sensory ganglia. Neuroscience 2013; 250: 622-31.
299
Sun G, Ren Q, Bai L, Zhang L. Phoenixin-20 suppresses lipopolysaccharide-induced inflammation in dental pulp cells. Chem Biol Interact 2020; 318108971
300
König M, Zimmer AM, Steiner H, Holmes F. Crawley.; Brownstein, M.; Zimmer, A. Pain responses, anxiety and aggression in mice deficient in pre-proenkephalin. Nature 1996; 383(6600): 535-8.
301
Rougeot C, Robert F, Menz L, Bisson JF, Messaoudi M. Systemically active human opiorphin is a potent yet non-addictive analgesic without drug tolerance effects. J Physiol Pharmacol 2010; 61(4): 483-90.
302
Wisner A, Dufour E, Messaoudi M, et al. Human Opiorphin, a natural antinociceptive modulator of opioid-dependent pathways. Proc Natl Acad Sci USA 2006; 103(47): 17979-84.
303
Ozdogan MS, Gungormus M, Yusufoglu S, Ertem SY, Sonmez C, Orhan M. Salivary opiorphin in dental pain: A potential biomarker for dental disease. Arch Oral Biol 2019; 99: 15-21.
304
Álvarez-Vásquez JL, Bravo-Guapisaca MI, Gavidia-Pazmiño JF, Intriago-Morales RV. Adipokines in dental pulp: Physiological, pathological, and potential therapeutic roles. J Oral Biosci 2022; 64(1): 59-70.
305
Lehr S, Hartwig S, Lamers D. Identification and validation of novel adipokines released from primary human adipocytes 2012.
306
Kritikou K, Totan A, Tanase M, Vinereanu A, Totan A, Spinu C. linca, R.; Miricescu, D.; Stanescu-Spinu J.; Greabu M. Biochemical Mapping of the Inflamed Human Dental Pulp. Appl Sci (Basel) 2021; 11(21): 10395.
307
Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med 2019; 25(12): 1822-32.
308
Liu L, Wang T, Huang D, Song D. Comprehensive analysis of differentially expressed genes in clinically diagnosed irreversible pulpitis by multiplatform data integration using a robust rank aggregation approach. J Endod 2021; 47(9): 1365-75.
309
Chen M, Zeng J, Yang Y, Wu B. Diagnostic biomarker candidates for pulpitis revealed by bioinformatics analysis of merged microarray gene expression datasets. BMC Oral Health 2020; 20(1): 279.
310
Fouad AF, Khan AA, Silva RM, Kang MK. Genetic and epigenetic characterization of pulpal and periapical inflammation. Front Physiol 2020; 11: 21.
311
Galicia JC, Henson BR, Parker JS, Khan AA. Gene expression profile of pulpitis. Genes Immun 2016; 17(4): 239-43.
312
Ricucci D, Loghin S, Siqueira JF. Correlation between clinical and histologic pulp diagnoses. J Endod 2014; 40: 1932-9.
313
Laine M, Ventä I, Hyrkäs T, Ma J, Konttinen YT. Chronic inflammation around painless partially erupted third molars. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2003; 95(3): 277-82.
314
Marciani RD. Is there pathology associated with asymptomatic third molars? J Oral Maxillofac Surg Off J Am Assoc Oral Maxillofac Surg 2012; 70(9)(Suppl. 1): S15-9.
315
Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal diseases. Lancet Lond Engl 2005; 366(9499): 1809-20.
316
Paloma de Oliveira B, Câmara AC, Aguiar CM. Prevalence of asymptomatic apical periodontitis and its association with coronary artery disease in a brazilian subpopulation. Acta Stomatol Croat 2017; 51(2): 106-12.
317
Karteva T, Manchorova-Veleva N. Biomarker for asymptomatic apical periodontitis in gingival crevicular fluid: aMMP-8. Eur J Dent 2020; 14(2): 239-44.
318
Buonavoglia A, Latronico F, Pirani C, Greco MF, Corrente M, Prati C. Symptomatic and asymptomatic apical periodontitis associated with red complex bacteria: clinical and microbiological evaluation. Odontology 2013; 101(1): 84-8.
319
Bennett DLH, Woods CG. Painful and painless channelopathies. Lancet Neurol 2014; 13(6): 587-99.
320
Nahorski MS, Chen YC, Woods CG. New Mendelian Disorders of Painlessness. Trends Neurosci 2015; 38(11): 712-24.
321
Simonetti M, Kuner R. Locus revealed: Painlessness via loss of NaV1.7 at central terminals of sensory neurons. Neuron 2021; 109(9): 1413-6.
322
Ziegler D, Landgraf R, Lobmann R, et al. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study). Diabetes Res Clin Pract 2018; 139: 147-54.
323
Testa G, Cattaneo A, Capsoni S. Understanding pain perception through genetic painlessness diseases: The role of NGF and proNGF. Pharmacol Res 2021; 169105662.
324
Goto M, Nakanaga K, Aung T, et al. Nerve damage in Mycobacterium ulcerans-infected mice: probable cause of painlessness in buruli ulcer. Am J Pathol 2006; 168(3): 805-11.
325
Amodio A, De Marchi G, de Pretis N, et al. Painless chronic pancreatitis. Dig Liver Dis 2020; 52(11): 1333-7.
326
Bhullar FA, Faghih M, Akshintala VS, Ahmed A, Lobnerc K. afgano, E.; Phillips, A.; Hart, P.; Ramsey, M.; Bick, B.; Kuhlmann, L.; Drewes, A.; Yadav, D.; Olesen, S.; Singh, V. Prevalence of primary painless chronic pancreatitis: A systematic review and meta-analysis. Pancreatology 2022; 22(1): 20-9.
327
Hollenbach M, Barresi L. Shedding light on painless chronic pancreatitis. Dig Liver Dis 2020; 52(11): 1331-2.