Next Article in Journal
LFA-1/ICAM-1 Adhesion Pathway Mediates the Homeostatic Migration of Lymphocytes from Peripheral Tissues into Lymph Nodes through Lymphatic Vessels
Previous Article in Journal
Synthesis and Characterization of Polyhydroxyalkanoate/Graphene Oxide/Nanoclay Bionanocomposites: Experimental Results and Theoretical Predictions via Machine Learning Models
Previous Article in Special Issue
HIV-1 Transcriptional Activator Tat Inhibits IL2 Expression by Preventing the Presence of Pol II on the IL2 Promoter
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nociceptor–Macrophage Interactions in Apical Periodontitis: How Biomolecules Link Inflammation with Pain

Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(8), 1193; https://doi.org/10.3390/biom13081193
Submission received: 20 June 2023 / Revised: 20 July 2023 / Accepted: 24 July 2023 / Published: 31 July 2023
(This article belongs to the Collection Feature Papers in Biological Factors)

Abstract

:
Periradicular tissues have a rich supply of peripheral afferent neurons, also known as nociceptive neurons, originating from the trigeminal nerve. While their primary function is to relay pain signals to the brain, these are known to be involved in modulating innate and adaptive immunity by initiating neurogenic inflammation (NI). Studies have investigated neuroanatomy and measured the levels of biomolecules such as cytokines and neuropeptides in human saliva, gingival crevicular fluid, or blood/serum samples in apical periodontitis (AP) to validate the possible role of trigeminal nociceptors in inflammation and tissue regeneration. However, the contributions of nociceptors and the mechanisms involved in the neuro-immune interactions in AP are not fully understood. This narrative review addresses the complex biomolecular interactions of trigeminal nociceptors with macrophages, the effector cells of the innate immune system, in the clinical manifestations of AP.

1. Introduction

Apical periodontitis (AP) is an inflammatory disease of periradicular tissue caused by oral opportunistic bacteria and their toxins retained in the apical regions of the periodontium [1,2]. Rich in blood vessels and nerve fibers, the periradicular tissue is composed of fibroblasts, epithelial cells, odontoblasts, cementoblasts, osteoblasts, osteoclasts, macrophages, and undifferentiated stem cells that form the periodontal ligament (PDL) and alveolar bone [3,4]. An infected root canal system in the apical periodontium causes all these immune cells and precursor cells to interact with one another, thereby eliciting an immune response against pathogens and pathogen-associated molecular patterns (PAMPs) [5]. However, AP can persist asymptomatically, without common clinical symptoms such as pain and swelling, while afferent sensory neurons that typically relay pain continue to engage in immune responses at the molecular level [6,7].
Neurogenic inflammation (NI) is set in motion when neuropeptides such as Substance P (SP) and Calcitonin Gene-Related Peptide (CGRP), among others, released by activated nociceptors act on resident immune cells and endothelial cells, causing vascular changes resulting in an influx of migrating immune cells to an inflamed tissue. To facilitate their function in NI, endothelial cells (vascular cells), mast cells, dendritic cells, T and B lymphocytes, and macrophages express cell-surface receptors for neuropeptides released by nociceptors [8,9]. Neuropeptides activate immune cells which release proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, nerve growth factor (NGF), and prostaglandin E2 (PGE2) that are in turn known to activate sensory neurons [10,11]. Neuropeptides also play a role in regulating anti-inflammatory roles of immune cells. For example, CGRP has been shown to inhibit the production of pro-inflammatory cytokines and enhance the expression of a regulatory phenotype in activated macrophages [12]. This reflects on the pleiotropic nature of neuropeptides that are capable of modulating immune response.
It is important to note that neurogenic inflammation is caused by the activation of peripheral nociceptors, resulting in inflammatory events in tissues that involve host innate and adaptive immune systems. Neuroinflammation, however, describes a localized host immune response to inflammation specifically in the brain and spinal cord [13,14].
Of the myriad cellular interactions in periradicular tissues, the cross-talk between macrophages and sensory neurons stands out because macrophages constitute up to 46% of inflammatory cells in AP [7]. This review aims to provide a comprehensive understanding of the temporal interactions between sensory neurons and macrophages while highlighting the role of nociceptors in different clinical manifestations of chronic AP.

2. Overview of Macrophages in Apical Periodontitis

Inflammation is the physiological response to injury, stress, or infection. In terms of the immune cells involved, inflammation is the state in which innate and adaptive cells are activated, undergo differentiation, and migrate to resolve inflammation while restoring tissue homeostasis. A rapid host immune response is capable of resolving acute inflammation. On the other hand, prolonged exposure to triggers such as damage-associated molecular patterns (DAMPs), PAMPS, and pathogens results in a state of chronic inflammation that sees different immune and non-immune cells in action [15].
Acute AP is marked by the presence of periapical abscesses, while periapical granulomas are typically a sign of chronic AP, typically diagnosed via the radiographic visualization of rarefying osteitis in periapical tissues. AP is dynamic in nature because of the different immune cell types participating in different stages of AP. This dynamic nature can be observed via the progressive change in the distribution of immune cells in the periapical inflammatory infiltrate [16,17]. Immune cells can also undergo polarization to different phenotypes in order to facilitate the host immune response in disease or wound-healing [18,19].
Physiologically, the PDL has a key role in responding to microbes located within the pulp space. The PDL is a highly vascularized tissue, with up to ten times higher vascular volume than other fibrous connective tissues [20]. The blood vessels in PDL primarily participate in tooth support and shock absorption [21]. The activation of immune cells such as macrophages leads to the release of proinflammatory cytokines that increase the expression of cellular adhesion molecule (CAM-1) in endothelial cells, thereby promoting the local adhesion of monocytes and leukocytes [18,22]. This local response, combined with vasodilation caused by initial inflammation response, leads to the active infiltration of monocytes to the site of inflammation, where growth factors, pro-inflammatory cytokines, and microbial products induce cellular differentiation [23,24]. Once at the site of inflammation, these circulatory monocytes can be differentiated into macrophages or dendritic cells [25].
Toll-like receptor-4 (TLR4) is a sensory receptor predominantly expressed by human myeloid cells (monocytes and granulocytes) that recognize PAMPs such as lipopolysaccharides (LPS) expressed by Gram-negative bacteria [26]. Activated TLR4 recruits adaptor proteins that undergo phosphorylation. Myeloid differentiation primary response 88 (MyD88), Interleukin-1 receptor-associated kinase (IRAK), and tumor necrosis factor receptor-associated factor-6 (TRAF-6) are examples of such adaptor proteins. Phosphorylated adaptor proteins activate Nuclear Factor kappa B (NFκB) and mitogen-activated protein kinase (MAPK) signaling pathways, resulting in the expression of pro-inflammatory cytokines [27]. Accessory molecules such as myeloid differentiation 2 (MD2), cluster of differentiation (CD)36, and CD14 can expedite TLR4 signaling [28]. Macrophages are also antigen-presenting cells that activate members of the adaptive immunity, effectively mounting an adaptive immune response.
Macrophages are known to play an active role in resolving inflammation [29]. Activated macrophages can polarize into functional phenotypes of classically activated M1, or alternatively activated M2, to bring about pro- and anti-inflammatory effects, respectively. M1 macrophages produce pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, which stimulate the production of tissue-damaging proteases and prostaglandins while mediating bone resorption. M1 macrophages also produce chemokines such as C-X-C motif chemokine ligand (CXCL)9 and CXCL10 that attract members of the adaptive immunity to the site of inflammation [30,31]. M2 macrophages are known to produce anti-inflammatory cytokines Transforming Growth Factor (TGF)-β and IL-10, which downregulate inflammation and immune response [32,33]. Their phagocytic capabilities help clear apoptotic cells and debris in healing tissues [5,34].

3. Role of Macrophages in Alveolar Bone Resorption

Resident macrophages in periodontal tissues secrete inflammatory cytokines and chemokines in response to pathogens. Qualitative and quantitative analyses have identified macrophages as the major constituent immune cell type in periapical granulomas [35]. This can be partly attributed to the vasodilation caused by the initial immune response, which in turn leads to the active infiltration of monocytes to the site of inflammation, wherein the growth factors, pro-inflammatory cytokines, and microbial products would induce differentiation to macrophages [24,36].
Alveolar bone resorption (loss) is a notable radiographic indication of chronic AP. The alveolar bone functions to hold teeth in the socket while absorbing any force applied to the teeth during biting and chewing. Subject to varying degrees of mechanical forces, the alveolar bone undergoes repeating cycles of bone formation and resorption. In a state of proinflammation, this equilibrium tends towards bone resorption, resulting in the loss of both mineral and organic components of the alveolar bone. The close association of macrophages with different stages of AP implicates these cells in the development of alveolar bone resorption. Macrophages were observed near osteoblasts in rat models of AP during the initial stages of bone resorption, while no macrophages were seen near the surfaces undergoing bone formation [19]. However, a study comparing data from immunohistochemical analyses of human specimens with clinical and radiological data reported no significant difference in the presence of macrophages between periapical granulomas and cysts [16]. Previous studies have highlighted disparities in the clinical presentation of AP [16,37]. Thus, a better understanding of the mechanisms underlying bone modeling and remodeling will help in identifying molecular pathways linking macrophages to alveolar bone resorption.
Osteoclastogenesis, or the development of mature osteoclasts (bone resorptive cells) from precursor cells, is mediated by receptor activator of nuclear factor kappa-β ligand (RANKL) [38]. RANKL is a cytokine produced by osteoblasts (which synthesize bone matrix and differentiate into osteocytes [39]) and stromal cells that binds to its receptor, RANK, to initiate the differentiation of osteoclast precursor cells [40]. This process is regulated by Osteoprotegerin (OPG), a decoy receptor of RANKL that is primarily produced by osteoblasts [41]. Hence, the RANKL-RANK-OPG trio are the main players in bone modeling and remodeling pathways [42]. GM-CSF (Granulocyte-Macrophage Colony-Stimulating Factor) is a cytokine that induces proliferation and maturation in macrophages, and it has been found to promote fusion of prefusion osteoclasts in the presence of RANKL, enabling bone resorption under pathologic and inflammatory conditions [43,44]. Besides GM-CSF, macrophages produce an array of inflammatory mediators in response to pathogens or microbial toxins that have been linked to bone resorption in AP [45]. IL-1 is one such proinflammatory cytokine that is produced by activated macrophages, which promote bone loss by enhancing RANKL expression by stimulating the synthesis of PGE2 in osteoblasts [46,47]. In addition, IL-1 is involved in the survival of osteoclasts and is known to induce multinucleation of osteoclasts, which is a hallmark of cell maturation [48]. Macrophages produce TNF-α, a proinflammatory cytokine that inhibits the differentiation and production of matrix proteins in osteoblasts [49,50]. TNF-α was found to play a role in osteoclastogenesis when increased RANKL mRNA expression was observed in osteocytes (which modulate bone remodeling [45]) that were cultured in vitro with exogenous TNF-α. This study observed an increased percentage of RANKL-positive osteocytes in mice injected with TNF-α [51].

4. Sensory Neuronal Activation in Apical Periodontitis

Periradicular tissues have an abundant supply of neurons that originate in the trigeminal ganglion (TG) [52]. Research groups in the 1970s identified nociceptive afferents with different activation thresholds and conduction velocities in the PDL [53]. Immunocytochemical analysis identified NGF-positive sensory nerves in periodontal tissues from rats [54], while ultrastructural analyses of the human PDL using electron microscopy identified Ruffini nerve endings, free nerve endings, and lamellated corpuscles [55]. Electrophysiological studies performed on cat models helped characterize trigeminal nerve endings in the PDL into low-threshold mechanoreceptors sensitive to mechanical load and vibration (A-β fibers) and nociceptors sensitive to pressure, temperature, and chemical irritants (A-δ, C-fibers) [56,57]. A-β fibers have large cell bodies, and these are myelinated [58,59]. A-δ fibers have medium-sized cell bodies, and C-fibers are considered the smallest [58,59,60]. Myelinated A-δ fibers have higher conduction velocities, resulting in a sharp pain sensation compared to the unmyelinated, slow C-fibers, which relay a delayed and dull, ache-like pain [58,61].
Studies implicating sensory neurons in inflammation have come a long way since the classic studies that concluded that neurogenic inflammation is based on axon reflexes [62,63]. While this theory did not pan out, some of the earlier works were successfully able to link activated sensory neurons to inflammatory phenomena such as vasodilation, setting stage for further studies [64,65]. As with the classical inflammatory response, neurogenic inflammation has also been associated with tissue regeneration [66]. It was reported that sensory denervation in rats, with pulp exposure to oral environment, showed the loss of pulpal tissue, while innervated teeth showed extensive neural sprouting and formation of reparative dentin and osteodentin [66].
Specialized surface receptors expressed by nociceptors play a crucial role in the cascade of events that led to NI (Figure 1). Transient receptor potential (TRP) channels are a class of integral membrane proteins that function as signal transducers in sensory neurons. These are polymodal in nature and can be activated by intracellular and/or extracellular mechanical, thermal, chemical, and osmotic pressure gradients. Based on sequence similarity, several classes of TRP channels exist. The TRPV subfamily (vanilloid subtype) has six members, of which TRPV1 has garnered attention for its role in the detection of noxious physical and chemical stimuli [67,68]. TRPA1 is the sole member of the TRPA subfamily (ankyrin subtype) [69,70]. The detailed 3D structure of the TRPA1 receptor was previously characterized by Cao et al. and Liao et al. [71,72]. Such studies made it possible to study the conformational changes that occur when TRPA1/TRPV1 receptors are activated. Their activation results in an active influx of extracellular calcium ions, which mediate action potentials in neurons and trigger exocytosis of CGRP and SP from large dense core vesicles (LDCVs) that involve members of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) family of proteins [73,74]. Biologically active neuropeptides are derived from their precursor peptides that are cleaved in the endoplasmic reticulum, after which these pro-peptides undergo post-translational modifications to form active versions that are stored in LDCVs prior to release from the sensory nerve endings [75,76]. A-δ and C fibers express TRPV1 and TRPA1 channels, and hence, these channels are associated with chemical and thermal nociception [77]. Knockout models of TRPV1 and TRPA1 highlight the role of these receptors in eliciting host immune response besides nociception. Non-neuronal cells such as endothelial cells, fibroblast, odontoblasts, and immune cells, including mast cells, monocytes, macrophages, and dendritic cells, also express TRPV1 and TRPA1 [77,78,79]. In these cells too, TRPV1 and TRPA1 exhibit functional responses to similar triggers as seen in neurons; however, it is not clear yet if the sensitivity thresholds of these receptors in non-neuronal cells are similar to those in neurons [77,80].
Toll-like receptors (TLRs) are another class of surface receptors that have been identified on nociceptors. The nociceptor expression of TLR4 highlights its role in the host immune response to bacterial infections [81,82]. Peripheral sensory neurons showed the colocalized expression of TLR4 and CD14, indicating the capability of the direct detection of PAMPs [82]. Besides immunological implications, the LPS binding of TLR4 on trigeminal neurons was shown to increase sensitization of TRPV1 channels, leading to increased expression of CGRP, which suggests a possible mechanism for pain associated with bacterial infections [83,84,85]. Similar findings were reported in an independent study that demonstrated a concentration-dependent effect of LPS on TRPV1 in trigeminal neurons [83].
Different bacterial strains have been shown to evoke different degrees of nociceptor response [86]. Gram-positive bacteria operate through mechanisms that are distinct from those triggered by Gram-negative bacteria [87]. S. aureus utilize N-formylated peptides and pore-forming toxin α-hemolysin to instigate pain in mice [87]. This study also showed a strong correlation between the bacterial load of live S. aureus and pain expressed by mice. Such studies provide insight into the different mechanisms that may be involved in eliciting pain in bacteria-mediated diseases such as AP, where both Gram-negative and Gram-positive strains play active roles in disease pathogenesis [2,88].

5. Role of Sensory Neuropeptides in Pain and Inflammation

Neuropeptides are considered the primary mediators of neurogenic inflammation. A-δ and C-fibers are peptidergic in nature, meaning these release neuropeptides mediating NI in AP [89,90] (Table 1). CGRP and SP are the principal neuropeptides involved in AP.
CGRP was discovered in 1982 by Amara et al. [135]. This 37-residue peptide was found to be expressed in the central and peripheral nervous systems, promoting sensory, motor, and autonomic functions in the brain [136]. CGRP has two isoforms, α-CGRP and β-CGRP, which have over 90% similar homology and share similar biological effects in humans [75]. CALC I gene undergoes alternate splicing to generate α-CGRP, while β-CGRP is a product of CALC II gene transcription. While α-CGRP is predominant in the central and peripheral nervous systems, β-CGRP is found mainly in the enteric nervous system [75]. Vasodilatory effects of human and rat CGRP were discovered in 1985, effectively linking CGRP to hyperemia [137]. The development of CGRP antagonists helped understand the role of CGRP in migraines, linking neuropeptides with pain pathways in the orofacial region and in other tissues [138]. Recognizing the ability of sensory nerves to produce and release CGRP led to the discovery of different stimuli that can activate neural cells to release this neuropeptide and the pathways that lead to the production and release post-activation [139].
The CGRP receptor molecule is a G protein-coupled receptor and is composed of three components: receptor activity modifying protein 1 (RAMP1), Calcitonin-like receptor (CLR), and receptor component protein (RCP). All three components are required to form a fully functional CGRP receptor (CGRP-R). Upon activation by binding with CGRP, CGRP-R may couple with G protein α subunit, increasing the cyclical adenosine monophosphate (cAMP) levels, which activate protein kinase A (PKA). This can result in the phosphorylation of cAMP response-element-binding (CREB) protein leading to gene transcription or the activation of extracellular signal-regulated kinases (ERK) pathway within a neural cell [139]. This signaling pathway can also result in the opening of ATP-sensitive K+ channels, which can lead to vasodilation [98].
Substance P (SP) is an undecapeptide that was first discovered in 1931 by Von Euler and Gaddum [140]. SP, neurokinin A (NKA), neuropeptide K (NPK), and neuropeptide γ (NPγ) are tachykinin neuropeptides encoded by the TAC1 gene; alternate RNA splicing and differential post-translational processing result in the expression of specific tachykinin peptides [141]. SP is best known for its role as a modulator of pain perception by altering cellular signaling pathways using G protein-coupled receptors called neurokinin 1 receptor (NK-1R) that can act through the cAMP secondary messenger system or inositol triphosphate (IP3)-diacylglycerol (DAG) system [108]. These downstream signaling pathways result in the expression of pro-inflammatory cytokines. SP can also exhibit autocrine effects on neural cells expressing NK-1R in a desensitizing process that results in the internal phosphorylation of NK-1R and recycling via endocytosis and acidification in endosomes [108].
CGRP and SP are known to colocalize, and these have been studied to assess the degree of pain experienced by patients with symptomatic AP [142]. Affirming their role in oral diseases, several studies have reported increased levels of CGRP and SP in the sites of inflammation in the dental pulp [143,144], periodontal ligament [145,146], and saliva and gingival crevicular fluid [111,147,148]. The levels of neuropeptides in these environments are correlated with the degrees of inflammation, confirming their role in inflammatory mechanisms [111,113,144].
CGRP and SP play vital roles in modulating bone resorption. In murine AP models, TRPV1-positive sensory neurons were shown to regulate osteoclastogenesis via CGRP signaling [104]. On the other hand, SP was found to up-regulate RANKL/OPG ratio in a rat model of periodontitis, promoting RANKL-induced osteoclast differentiation [115].
SP and CGRP have also been shown to mediate tissue repair. The sprouting of CGRP-positive fibers in odontoblast layers after injury induced in the cervical dentin of rats resulted in the formation of reparative dentin [149]. SP released by stimulated peripheral nerves increased blood flow to the dental pulp, thereby enabling tissue healing [113,116]. Hence, SP and CGRP can be considered immunomodulators that can have pro- and anti-inflammatory effects depending on molecular cues present in the tissue microenvironment.

6. Neuro-Immune Interactions Modulate Inflammation and Healing in AP

Evidence of a functional link between sensory neurons and immune cells comes from the proximity of sensory nerve endings and immune cells. This is corroborated by the expression of neuropeptide-specific receptors on immune cells and cytokine receptors and receptors capable of recognizing microbial pathogens on trigeminal nociceptor neurons [8,150] (Figure 1 and Figure 2).
In vitro studies have confirmed the expression of CGRP-R and NK-1R in murine and human macrophages, implying that neuropeptides CGRP and SP influence the activities of these immune cells [99,151]. Pro-inflammatory cytokines IL-1, IL-4, and interferon (IFN)-γ were also shown to induce NK-1R expression in macrophages [152,153]. Goldman et al. were among the first to conduct in vitro studies on peritoneal macrophages, showing enhanced phagocytosis when exogenous SP was applied to these cells [154]. The SP-mediated activation of macrophages was discovered when exposure to SP increased levels of eicosanoids in macrophages [155]. SP also acts as a chemoattractant for monocytes and macrophages, first demonstrated in vitro using human monocytes and peritoneal macrophages from guinea pigs [156,157]. Evidence of TNF-α, IL-1, and IL-6 release in macrophages in response to SP was also previously recorded [158]. In contrast, CGRP was found to actively prevent the activation of monocytes and macrophages [102,103]. Exogenous CGRP reduced proliferation in peripheral blood mononuclear cells and inhibited macrophage production of hydrogen peroxide, which is considered a classic response to inflammation [102,103].
Early identification of CGRP immunoreactive (CGRP-IR) nerve fibers in the PDL led to increased interest in studying their response to different stimuli. Dynamic changes were observed in CGRP-IR nerve densities surrounding blood vessels in PDL during tooth movement in rat molars [159]. Wakisaka et al. were among the first to study the distribution of SP-positive nerve fibers in rat PDL [160]. However, when immunoreactions for CGRP, SP, and neuropeptide Y (NPY) were evaluated in dental pulp, periodontal ligament, and gingiva in cats, denser innervation in the pulp was reported, and these nerves were found to be more immunoreactive to CGRP than to SP [161]. A similar trend was observed by the same group in a ferret model, affirming a similar abundance in sensory nerve innervation across different species [162]. Hence, several subsequent studies focused on studying CGRP-positive neurons. Kimberly and Byers induced pulpitis that led to periapical lesions in rat models and monitored changes in CGRP-IR nerves to inflammation for 35 days [163]. They observed that while inflammation initially caused nerve damage, increased neural sprouting was seen along the wound border. Even as inflammation advanced in severity and transitioned from the pulp to the periapical regions, CGRP-IR nerves sprouted extensively in both tissues. Independent studies reported similar patterns of neural sprouting in rat models, linking the response of pulpal innervation to inflammation and the induction of periapical lesions [164].
Dynamic changes in the distribution of sensory nerves during acute and chronic AP have drawn attention to understanding how these specialized neurons may modulate disease progression. Several studies measured increased levels of neuropeptides in saliva and gingival crevicular fluid (GCF) in animal models and in patients diagnosed with AP [165,166]. Similarly, investigations on the distribution patterns of neuropeptides in periodontitis suggested the potential of using neuropeptides as biomarkers for the early detection of periodontitis [167,168]. A significant increase in SP-like immunoreactivity was measured in GCF collected from periodontitis sites of volunteers [169]. Studies also found a significant correlation between SP and proinflammatory IL-1β, TNF-α, IL-8, and chemoattractant MCP-1 (monocyte chemoattractant protein-1) in GCF from patients with periodontitis [170]. CGRP exhibited a different profile in comparison; reduced CGRP immunoreactivity was observed in periodontitis sites compared to healthy periodontium [97]. Further analysis showed that carboxypeptidase activity in GCF resulted in a rapid breakdown of CGRP; however, this did not affect the levels of SP in GCF [171].
Macrophages express functional CGRP-R, i.e., receptors of CGRP. CGRP activation led to the proliferation and differentiation of murine macrophages into osteoclast-like cells via a cAMP/PKA-dependent signaling pathway [172]. Murine bone-marrow-derived macrophages (BMDMs) showed increased IL-6 expression as a response to CGRP activation, which led to speculations about the role of BMDMs in the negative inhibition of B cell development [173]. Similar findings were reported in rat models that demonstrated reduced IL-6 expression in invading macrophages upon perineural injection of a CGRP antagonist [100]. In vitro studies have shown that LPS-induced expression of pro-inflammatory cytokines, including IL-1β, IL-6, contributes to CGRP expression in macrophages [99].
Primarily produced by sensory neurons in the periodontal tissues, NGF is a neurotrophic factor that plays a critical role in neurogenesis, promoting the growth, differentiation, and survival of neurons [174,175]. Nerve Growth Factor-Tropomyosin receptor kinase A (NGF/TrkA) signaling suppresses CGRP production in monocytes as well as RAW264.7 macrophages [99]. In periodontal tissues that have a rich sensory nerve supply, NGF can induce increased neural sprouting and innervation under inflammation. NGF also controls the expression and release of neuropeptides in sensory neurons. NGF-mediated neural responses have direct implications on the host tissue response to inflammation [176,177]. NGF has been shown to induce monocyte differentiation in macrophages, thereby enhancing inflammatory effects [178]. Non-neural cells such as fibroblasts and osteoblasts can produce NGF in periodontal tissues, and several non-neural cell types express TrkA, which is a high-affinity NGF receptor molecule [179,180]. Hematopoietic stem cells express TrkA and produce their own NGF to support differentiation in these cells. This expression is maintained so that neutrophils, mast cells, and monocytes that arise from these stem cells also express TrkA. An interesting finding was that the expression of TrkA in monocytes and macrophages had functional implications that increased when these cells were stimulated with bacterial toxins [178,181]. For example, TrkA-stimulated human monocytes showed an upregulation of anti-apoptotic genes [182]. Another study showed that monocytes production of CGRP was reduced in response to LPS when NGF was neutralized [183]. TrkA-dependent ruffling of human-monocyte-derived macrophage cell membrane, induced by NGF, was found to increase phagocytic activity [184]. NGF also induced increased the macrophage expression of matrix metalloproteinase (MMP)-9 in this study. Although the functions and release mechanisms of neurotrophins in immune cells remain elusive, current knowledge substantiates the role of sensory-neuron-derived NGF in macrophage functions under inflammation.
SP was found to bind NK-1R receptors on murine macrophages and activate protein kinase C (PKC) or phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), which triggers nuclear factor kappa-light-chain-enhancer of activated B cells transactivation and chemokine response via ERK1/2 and p38 MAPK signaling pathways [185,186]. Both resulted in the expression of the chemokines macrophage inflammatory protein-2 (MIP-2) and MCP-1. MIP-2 is a potent chemoattractant for neutrophils, and MCP-1 regulates monocyte migration and infiltration [187,188]. Macrophages produce SP in response to NF-κB activation, linking this phenomenon with the pro-inflammatory M1 macrophage phenotype. The administration of exogenous SP has been found to increase IL-10 expression in rat models, creating conducive environment for the activation of anti-inflammatory M2 macrophages, which can also produce SP [189,190]. SP was found to induce hemeoxygenase-1 (HO-1), an antioxidant, in LPS-stimulated murine macrophages in a dose-dependent manner [191]. HO-1 is expressed in response to oxidative stress and functions to suppress inflammation by inducing macrophage polarization in favor of an M2 anti-inflammatory phenotype [192]. SP also induced M2-like macrophages by activating PI3K/Akt in murine macrophages [193]. Hence, there is evidence of both pro- and anti-inflammatory effects of SP, which warrants detailed studies in AP.

7. Association of Pain with Nociceptor-Macrophage Cross-talk in AP

Pain is a natural response to tissue damage, and current investigations highlight a close association of this sensation with nociceptor–immune-cell interactions [194]. Several aspects can contribute to the presentation of pain in endodontic infections, including the volume of root canal space, the presence of pathogen diversity, the virulence of pathogenic load, the state of the immune system in an individual, and lesion dynamics, among others [195]. Despite standardized systems such as Numeric Rating Scale (NRS-11) and periapical index (PAI), which can correlate pain with clinical presentations in AP, these cannot be used to predict when and why pain is not experienced by patients with chronic AP [196,197]. Understanding pain perception from the perspectives of periapical inflammation on a molecular level may help improve diagnosis and develop better treatment plans and strategies.
Immune cells modulate nociceptor activity using cytokines and chemokines, and nociceptors in turn modulate functions of immune cells by expressing neuropeptides, resulting in a dynamic, bi-directional cross-talk between these two cell entities during tissue injury and inflammation [198] (Figure 3). Nociceptors express receptor molecules that are activated by PAMPs, DAMPs, and mediators released by immune cells. Neutrophils, mast cells, and macrophages have close encounters with nociceptive nerve terminals, and hence, mediators produced by these immune cells can induce pain sensitization and influence chronicity of pain. Macrophages produce TNF-α, IL-1β, IL-6, PGE2, and Leukotriene B4 (LTB4), covering a range of cytokines, growth factors, and lipids that bind to specific receptors on nociceptor neurons and result in direct pain sensitization or engage ion channels to increase neuronal cell excitation which then causes pain sensitization [194]. Such pain sensitization translates to hyperalgesia or allodynia in AP.
Hyperalgesia and allodynia refer to increased nociceptive sensitivity in peripheral nerves. While both are caused by the reduced activation threshold of nociceptor neurons, hyperalgesia is caused by repeated or intense exposure to noxious mechanical or thermal stimuli, and allodynia is associated with non-toxic mechanical or thermal stimulation. A comparison of maximal bite force in contralateral, non-inflamed teeth in patients with acute periradicular periodontitis against teeth from healthy individuals reported allodynia in the contralateral teeth of acute AP patients. This was attributed to central sensitization, which refers to an increase in sensitivity in neurons in the central nervous system (CNS) [199,200]. Owatz et al. proposed that central sensitization may evoke mechanical allodynia in pulpal nociceptors independently of the local activation of the nociceptors in pulpal tissue [201]; this is yet to be confirmed. Studies have found correlations between nociceptive pain and neuropeptide CGRP [138,202]. Mechanical allodynia was reported when CGRP was injected into the hind paw skin of mice [203]. The intradermal injection of CGRP antagonist after 1 h of capsaicin injection resulted in a partially reversed hyperalgesia and mechanical allodynia in rats, confirming the role of CGRP in maintaining nociceptive sensitivity [204].
De França et al. evaluated M1 and M2 macrophage populations in human radicular cysts and periapical granulomas the presence of both are classic markers of apical periodontitis [205]. The authors also correlated pain with macrophage subpopulations in samples tested; a higher prevalence of macrophages was observed in areas with higher densities of microorganisms [205]. Based on sample collection, more cases with radicular cysts were symptomatic compared to periapical granulomas, meaning this trend could be attributed to the pro-inflammatory profile of cysts that were assumed to have increased levels of IL-1, IL-6, and PGE2, all of which are released by macrophages in response to microbial infection and have been identified in causing painful symptoms. However, periapical granulomas were characterized by pulsating pain in this study, and this was linked to higher inflammatory infiltrate that also had higher amounts of CD68+ cell populations. A correlation between macrophages and CGRP, known to initiate and maintain pain, was reported when the increased presence of CGRP-immunoreactive fibers was observed along with an increased number of resident macrophages in rat models of AP [206]. These studies have directly or indirectly highlighted an important link between macrophages and pain sensations in AP. There is, however, a lack of consensus that successfully links macrophages and pain in AP progression from radicular cysts to periapical granulomas [205].
A recent study found correlations between the immunoexpression of MMPs and the presence of pain in patients with AP; periapical granulomas and periapical cysts in teeth extracted from patients who were evaluated for the presence of pain showed high expression levels of MMP-2 [207]. MMPs are a class of enzymes that participate in the degradation of matrix proteins and bone, contributing to the processes of tissue degradation and remodeling. Categorized into different classes such as collagenases, gelatinases, stromelysins, and others, MMPs are also capable of cleaving cytokines and chemokines, thereby modulating immune response. MMP-1, produced by activated macrophages, has been found in increased concentrations in rat and mouse models of periapical lesions [208,209]. MMP-2, MMP-3, MMP-8, MMP-9, and MMP-13 have also been identified in human periapical lesions; of these, macrophages have been shown to express all of these in AP [210,211,212]. Although their mechanism of action is not fully understood, it is known that, in response to different cues, MMPs are secreted by different cell types into the extracellular matrix, where they are activated proteolytically and their actions are regulated by endogenous inhibitors called tissue inhibitors of metalloproteinases (TIMPs) [213,214]. In the context of AP, pro-inflammatory cytokines TNF-α and IL-1β are known to activate MMP-1 and MMP-2; macrophages are a major source of these cytokines [213,214]. Stimulating pain in the upper central incisor of human volunteers resulted in elevated levels of MMP-8 in the gingival crevicular fluid [112]. This study observed an increase in SP levels in the GCF of stimulated teeth, thereby correlating neuronal activation associated with pain with immune activation.

8. Conclusions

The discovery of neurogenic inflammation highlighted the role of nociceptors in the immune response. The dental pulp and periapical tissues are rich in sensory nerves, and nociceptors are now known to activate key elements of the innate immune system besides sending pain signals to indicate infection. Neuropeptides and inflammatory mediators released by both nociceptors and immune cells regulate neuro-immune cross-talk. Animal models and in vitro studies have led to a deeper understanding of neuro-immune cross-talk; however, we are yet to understand how microbial pathogens, immune cells, and nociceptors interact with each other resulting in the modulation of pain and inflammation in AP. Chronic AP is especially challenging to treat or manage because it is usually asymptomatic, persisting in a state of continued inflammation with varying presenting symptoms. Macrophages play a critical role in sensitizing and desensitizing nociceptors, leading to hyperalgesia, allodynia, and central sensitization in patients with AP. A comprehensive insight into the molecular mechanisms that connect neuro-immune interactions, inflammation, and pain is essential to understand periapical host response in symptomatic and asymptomatic AP while offering significant translational potential.

Author Contributions

A.K. contributed to the conception of the review. N.M. and A.K. wrote the manuscript with supervision from A.K. Both authors provided critical feedback and helped to shape the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The authors gratefully acknowledge the financial support from the Natural Sciences and Engineering Research Council of Canada (Discovery grant) (AK-RGPIN-2020-05844), Canada Research Chair program (AK), and the Dr. Lloyd and Mrs. Kay Chapman Chairship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A. List of Acronyms

APApical periodontitis
BMDMsBone-marrow-derived macrophages
CAM-1Cellular adhesion molecule 1
cAMPCyclical adenosine monophosphate
CDCluster of differentiation
CGRPCalcitonin Gene-Related Peptide
CGRP-IRCGRP immunoreactive
CLRCalcitonin-like receptor
CNSCentral nervous system
CREBcAMP response element binding
CXC-LChemokine C-X-C motif chemokine ligand
DAGDiacylglycerol
DAMPsDamage-associated molecular patterns
ERKsExtracellular signal-regulated kinases
GCFGingival crevicular fluid
GM-CSFGranulocyte-Macrophage Colony-Stimulating Factor
HO-1Hemeoxygenase-1
IFN-γInterferon-γ
IL-1Interleukin-1
IP3Inositol triphosphate
IRAKInterleukin-1 receptor-associated kinase
LDCVsLarge dense core vesicles
LPSLipopolysaccharides
LTB4Leukotriene B4
M1Classically activated macrophages
M2Alternatively activated macrophages
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein-1
MD2Myeloid differentiation 2
MIP-2Macrophage Inflammatory Protein-2
MMPsMatrix metalloproteinases
MQMacrophage
MyD88Myeloid differentiation primary response 88
NFκBNuclear Factor Kappa B
NGFNerve Growth Factor
NINeurogenic Inflammation
NKANeurokinin A
NPsNeuropeptides
NP γNeuropeptide γ
NPKNeuropeptide K
NPYNeuropeptide Y
NRS-11Numeric Rating Scale
OPGOsteoprotegerin
PACAPPituitary adenylate cyclase-activating peptide
PAIPeriapical index
PAMPsPathogen-associated molecular patterns
PDLPeriodontal ligament
PGE2Prostaglandin E2
PI3K/AktPhosphatidylinositol 3-kinase/protein kinase B
PKCProtein Kinase C
RANKLReceptor activator of nuclear factor kappa-β ligand
SNARESoluble N-ethylmaleimide-sensitive factor attachment protein receptor
SPSubstance P
TGTrigeminal ganglion
TGF-βTransforming growth factor-β
TIMPsTissue inhibitors of metalloproteinases
TLRToll-like receptor
TNF-αTumor necrosis factor-α
TRAF-6Tumor necrosis factor Receptor Associated Factor-6
TrkATropomyosin receptor kinase A
TRPTransient receptor potential
VIPVasoactive intestinal peptide

References

  1. Nair, P.N.R. On the causes of persistent apical periodontitis: A review. Int. Endod. J. 2006, 39, 249–281. [Google Scholar] [CrossRef] [PubMed]
  2. Korona-Glowniak, I.; Piatek, D.; Fornal, E.; Lukowiak, A.; Gerasymchuk, Y.; Kedziora, A.; Bugla-Płoskonska, G.; Grywalska, E.; Bachanek, T.; Malm, A. Patterns of Oral Microbiota in Patients with Apical Periodontitis. J. Clin. Med. 2021, 10, 2707. [Google Scholar] [CrossRef] [PubMed]
  3. Hassell, M.T. Tissues and cells of the periodontium. Periodontology 2000 1993, 3, 9–38. [Google Scholar] [CrossRef]
  4. Nanci, A.; Bosshardt, D.D. Structure of periodontal tissues in health and disease*. Periodontology 2000 2006, 40, 11–28. [Google Scholar] [CrossRef] [PubMed]
  5. Hussein, H.; Kishen, A. Local Immunomodulatory Effects of Intracanal Medications in Apical Periodontitis. J. Endod. 2022, 48, 430–456. [Google Scholar] [CrossRef] [PubMed]
  6. Jonasson, P.; Kvist, T. Diagnosis of apical periodontitis in root-filled teeth. Clin. Dent. Rev. 2018, 2, 15. [Google Scholar] [CrossRef] [Green Version]
  7. Li, F.-C.; Kishen, A. 3D Organoids for Regenerative Endodontics. Biomolecules 2023, 13, 900. [Google Scholar] [CrossRef]
  8. Chiu, I.M.; von Hehn, C.A.; Woolf, C.J. Neurogenic inflammation and the peripheral nervous system in host defense and immunopathology. Nat. Neurosci. 2012, 15, 1063–1067. [Google Scholar] [CrossRef] [Green Version]
  9. Assas, B.M.; Pennock, J.I.; Miyan, J.A. Calcitonin gene-related peptide is a key neurotransmitter in the neuro-immune axis. Front. Neurosci. 2014, 8, 23. [Google Scholar] [CrossRef] [Green Version]
  10. Kabata, H.; Artis, D. Neuro-immune crosstalk and allergic inflammation. J. Clin. Investig. 2019, 129, 1475–1482. [Google Scholar] [CrossRef] [Green Version]
  11. Crosson, T.; Roversi, K.; Balood, M.; Othman, R.; Ahmadi, M.; Wang, J.-C.; Seadi Pereira, P.J.; Tabatabaei, M.; Couture, R.; Eichwald, T.; et al. Profiling of how nociceptor neurons detect danger—New and old foes. J. Intern. Med. 2019, 286, 268–289. [Google Scholar] [CrossRef] [PubMed]
  12. Baliu-Piqué, M.; Jusek, G.; Holzmann, B. Neuroimmunological communication via CGRP promotes the development of a regulatory phenotype in TLR4-stimulated macrophages. Eur. J. Immunol. 2014, 44, 3708–3716. [Google Scholar] [CrossRef] [PubMed]
  13. Ji, R.-R.; Xu, Z.-Z.; Gao, Y.-J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014, 13, 533–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Matsuda, M.; Huh, Y.; Ji, R.-R. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J. Anesth. 2019, 33, 131–139. [Google Scholar] [CrossRef] [PubMed]
  15. Zehnder, M.; Belibasakis, G.N. On the dynamics of root canal infections—What we understand and what we don′t. Virulence 2015, 6, 216–222. [Google Scholar] [CrossRef] [Green Version]
  16. Azeredo, S.V.; Brasil, S.C.; Antunes, H.; Marques, F.V.; Pires, F.R.; Armada, L. Distribution of macrophages and plasma cells in apical periodontitis and their relationship with clinical and image data. J. Clin. 2017, 9, e1060–e1065. [Google Scholar] [CrossRef] [Green Version]
  17. Márton, I.J.; Kiss, C. Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis. J. Endod. 2014, 40, 155–163. [Google Scholar] [CrossRef]
  18. Nair, P.N.R. Pathogenesis of Apical Periodontitis and the Causes of Endodontic Failures. Crit. Rev. Oral Biol. Med. 2004, 15, 348–381. [Google Scholar] [CrossRef] [Green Version]
  19. Akamine, A.; Anan, H.; Hamachi, T.; Maeda, K. A histochemical study of the behavior of macrophages during experimental apical periodontitis in rats. J. Endod. 1994, 20, 474–478. [Google Scholar] [CrossRef]
  20. Sims, M.R. A model of the anisotropic distribution of microvascular volume in the periodontal ligament of the mouse mandibular molar. Aust. Orthod. J. 1987, 10, 21–24. [Google Scholar]
  21. Masset, A.; Staszyk, C.; Gasse, H. The blood vessel system in the periodontal ligament of the equine cheek teeth—Part II: The micro-architecture and its functional implications in a constantly remodelling system. Ann. Anat. Anat. Anz. 2006, 188, 535–539. [Google Scholar] [CrossRef] [PubMed]
  22. Graunaite, I.; Lodiene, G.; Maciulskiene, V. Pathogenesis of apical periodontitis: A literature review. J. Oral Maxillofac. Res. 2012, 2, e1. [Google Scholar] [CrossRef] [PubMed]
  23. Cekici, A.; Kantarci, A.; Hasturk, H.; Van Dyke, T.E. Inflammatory and immune pathways in the pathogenesis of periodontal disease. Periodontology 2000 2014, 64, 57–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-resident macrophages. Nat. Immunol. 2013, 14, 986–995. [Google Scholar] [CrossRef]
  25. Yang, J.; Zhang, L.; Yu, C.; Yang, X.-F.; Wang, H. Monocyte and macrophage differentiation: Circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark. Res. 2014, 2, 1. [Google Scholar] [CrossRef] [Green Version]
  26. Molteni, M.; Gemma, S.; Rossetti, C. The Role of Toll-Like Receptor 4 in Infectious and Noninfectious Inflammation. Mediat. Inflamm. 2016, 2016, 6978936. [Google Scholar] [CrossRef] [Green Version]
  27. Rayees, S.; Rochford, I.; Joshi, J.C.; Joshi, B.; Banerjee, S.; Mehta, D. Macrophage TLR4 and PAR2 Signaling: Role in Regulating Vascular Inflammatory Injury and Repair. Front. Immunol. 2020, 11, 2091. [Google Scholar] [CrossRef]
  28. Perros, F.; Lambrecht, B.N.; Hammad, H. TLR4 signalling in pulmonary stromal cells is critical for inflammation and immunity in the airways. Respir. Res. 2011, 12, 125. [Google Scholar] [CrossRef] [Green Version]
  29. Garlet, G.P.; Giannobile, W.V. Macrophages: The Bridge between Inflammation Resolution and Tissue Repair? J. Dent. Res. 2018, 97, 1079–1081. [Google Scholar] [CrossRef]
  30. House, I.G.; Savas, P.; Lai, J.; Chen, A.X.Y.; Oliver, A.J.; Teo, Z.L.; Todd, K.L.; Henderson, M.A.; Giuffrida, L.; Petley, E.V.; et al. Macrophage-Derived CXCL9 and CXCL10 Are Required for Antitumor Immune Responses Following Immune Checkpoint Blockade. Clin. Cancer Res. 2020, 26, 487–504. [Google Scholar] [CrossRef] [Green Version]
  31. Yao, Y.; Cai, X.; Ren, F.; Ye, Y.; Wang, F.; Zheng, C.; Qian, Y.; Zhang, M. The Macrophage-Osteoclast Axis in Osteoimmunity and Osteo-Related Diseases. Front. Immunol. 2021, 12, 664871. [Google Scholar] [CrossRef] [PubMed]
  32. Italiani, P.; Boraschi, D. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Front. Immunol. 2014, 5, 514. [Google Scholar] [CrossRef] [PubMed]
  33. Lappin, D.F.; Macleod, C.P.; Kerr, A.; Mitchell, T.J.; Kinane, D.F. Anti-inflammatory cytokine IL-10 and T cell cytokine profile in periodontitis granulation tissue. Clin. Exp. Immunol. 2001, 123, 294–300. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, L.; Hua, F.; Ding, W.; Ding, K.; Zhang, Y.; Xu, C. The correlation between the Th17/Treg cell balance and bone health. Immun. Ageing 2020, 17, 30. [Google Scholar] [CrossRef]
  35. Stern, M.H.; Dreizen, S.; Mackler, B.F.; Selbst, A.G.; Levy, B.M. Quantitative analysis of cellular composition of human periapical granuloma. J. Endod. 1981, 7, 117–122. [Google Scholar] [CrossRef]
  36. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.-A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef] [PubMed]
  37. Lukić, A.; Danilovic, V.; Petrović, R. Comparative immunohistochemical and quantitative analysis of inflammatory cells in symptomatic and asymptomatic chronic periapical lesions. Vojnosanit. Pregl. 2008, 65, 435–440. [Google Scholar] [CrossRef]
  38. Kawai, T.; Matsuyama, T.; Hosokawa, Y.; Makihira, S.; Seki, M.; Karimbux, N.Y.; Goncalves, R.B.; Valverde, P.; Dibart, S.; Li, Y.-P.; et al. B and T lymphocytes are the primary sources of RANKL in the bone resorptive lesion of periodontal disease. Am. J. Pathol. 2006, 169, 987–998. [Google Scholar] [CrossRef] [Green Version]
  39. Dallas, S.L.; Bonewald, L.F. Dynamics of the transition from osteoblast to osteocyte. Ann. N. Y. Acad. Sci. 2010, 1192, 437–443. [Google Scholar] [CrossRef] [Green Version]
  40. Pérez-Sayáns, M.; Somoza-Martín, J.M.; Barros-Angueira, F.; Rey, J.M.G.; García-García, A. RANK/RANKL/OPG role in distraction osteogenesis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2010, 109, 679–686. [Google Scholar] [CrossRef]
  41. Wada, T.; Nakashima, T.; Hiroshi, N.; Penninger, J.M. RANKL–RANK signaling in osteoclastogenesis and bone disease. Trends Mol. Med. 2006, 12, 17–25. [Google Scholar] [CrossRef]
  42. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch. Biochem. Biophys. 2008, 473, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ushach, I.; Zlotnik, A. Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage. J. Leukoc. Biol. 2016, 100, 481–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lee, M.S.; Kim, H.S.; Yeon, J.-T.; Choi, S.-W.; Chun, C.H.; Kwak, H.B.; Oh, J. GM-CSF Regulates Fusion of Mononuclear Osteoclasts into Bone-Resorbing Osteoclasts by Activating the Ras/ERK Pathway1. J. Immunol. 2009, 183, 3390–3399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hienz, S.A.; Paliwal, S.; Ivanovski, S. Mechanisms of Bone Resorption in Periodontitis. J. Immunol. Res. 2015, 2015, 615486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Gupta, G. Gingival crevicular fluid as a periodontal diagnostic indicator- II: Inflammatory mediators, host-response modifiers and chair side diagnostic aids. J. Med. Life 2013, 6, 7–13. [Google Scholar]
  47. Lee, Y.-M.; Fujikado, N.; Manaka, H.; Yasuda, H.; Iwakura, Y. IL-1 plays an important role in the bone metabolism under physiological conditions. Int. Immunol. 2010, 22, 805–816. [Google Scholar] [CrossRef] [Green Version]
  48. Jimi, E.; Shuto, T.; Koga, T. Macrophage colony-stimulating factor and interleukin-1 alpha maintain the survival of osteoclast-like cells. Endocrinology 1995, 136, 808–811. [Google Scholar] [CrossRef]
  49. Gilbert, L.; He, X.; Farmer, P.; Boden, S.; Kozlowski, M.; Rubin, J.; Nanes, M.S. Inhibition of Osteoblast Differentiation by Tumor Necrosis Factor-α*. Endocrinology 2000, 141, 3956–3964. [Google Scholar] [CrossRef]
  50. Lee, H.-L.; Yi, T.; Woo, K.M.; Ryoo, H.-M.; Kim, G.-S.; Baek, J.-H. Msx2 mediates the inhibitory action of TNF-α on osteoblast differentiation. Exp. Mol. Med. 2010, 42, 437–445. [Google Scholar] [CrossRef] [Green Version]
  51. Marahleh, A.; Kitaura, H.; Ohori, F.; Kishikawa, A.; Ogawa, S.; Shen, W.-R.; Qi, J.; Noguchi, T.; Nara, Y.; Mizoguchi, I. TNF-α Directly Enhances Osteocyte RANKL Expression and Promotes Osteoclast Formation. Front. Immunol. 2019, 10, 2925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Willis, R.D.; DiCosimo, C.J. The absence of proprioceptive nerve endings in the human periodontal ligament: The role of periodontal mechanoreceptors in the reflex control of mastication. Oral Surg. Oral Med. Oral Pathol. 1979, 48, 108–115. [Google Scholar] [CrossRef] [PubMed]
  53. Byers, M.R. Sensory innervation of periodontal ligament of rat molars consists of unencapsulated Ruffini-like mechanoreceptors and free nerve endings. J. Comp. Neurol. 1985, 231, 500–518. [Google Scholar] [CrossRef] [PubMed]
  54. Byers, M.R. Segregation of NGF receptor in sensory receptors, nerves and local cells of teeth and periodontium demonstrated by EM immunocytochemistry. J. Neurocytol. 1990, 19, 765–775. [Google Scholar] [CrossRef] [PubMed]
  55. Lambrichts, I.; Creemers, J.; van Steenberghe, D. Morphology of neural endings in the human periodontal ligament: An electron microscopic study. J. Periodontal Res. 1992, 27, 191–196. [Google Scholar] [CrossRef] [PubMed]
  56. Tabata, T.; Karita, K. Response properties of periodontal mechanosensitive fibers in the superior dental nerve of the cat. Exp. Neurol. 1986, 94, 469–478. [Google Scholar] [CrossRef]
  57. Karita, K.; Tabata, T. Response fields of the periodontal mechanosensitive units in the superior alveolar nerve of the cat. Exp. Neurol. 1985, 90, 558–565. [Google Scholar] [CrossRef]
  58. Tender, G.C.; Li, Y.-Y.; Cui, J.-G. Vanilloid receptor 1-positive neurons mediate thermal hyperalgesia and tactile allodynia. Spine J. 2008, 8, 351–358. [Google Scholar] [CrossRef]
  59. Christine, W.; Michael, G.H. Neuroanatomy, Touch Receptor; StatPearls Publishing: St. Petersburg, FL, USA, 2023. [Google Scholar]
  60. Mengel, M.K.C.; Jyväsjärvi, E.; Kniffki, K.-D. Identification and characterization of afferent periodontal C fibres in the cat. PAIN 1992, 48, 413–420. [Google Scholar] [CrossRef] [PubMed]
  61. Bender, I.B. Pulpal Pain Diagnosis—A Review. J. Endod. 2000, 26, 175–179. [Google Scholar] [CrossRef]
  62. Bayliss, W.M. On the origin from the spinal cord of the vaso-dilator fibres of the hind-limb, and on the nature of these fibres1. J. Physiol. 1901, 26, 173–209. [Google Scholar] [CrossRef] [PubMed]
  63. Lewis, T. The Blood Vessels of the Human Skin. Br. Med. J. 1926, 2, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bayliss, W.M. The Vaso-Motor System/by William M. Bayliss; Longmans, Green: New York, NY, USA, 1923. [Google Scholar]
  65. Langley, J.N. Antidromic action. J. Physiol. 1923, 57, 428–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Byers, M.R.; Taylor, P.E. Effect of Sensory Denervation on the Response of Rat Molar Pulp to Exposure Injury. J. Dent. Res. 1993, 72, 613–618. [Google Scholar] [CrossRef] [PubMed]
  67. Toledo Mauriño, J.J.; Fonseca-Camarillo, G.; Furuzawa-Carballeda, J.; Barreto-Zuñiga, R.; Martínez Benítez, B.; Granados, J.; Yamamoto-Furusho, J.K. TRPV Subfamily (TRPV2, TRPV3, TRPV4, TRPV5, and TRPV6) Gene and Protein Expression in Patients with Ulcerative Colitis. J. Immunol. Res. 2020, 2020, 2906845. [Google Scholar] [CrossRef]
  68. Jara-Oseguera, A.; Simon, A.S.; Rosenbaum, T. TRPV1: On the Road to Pain Relief. Curr. Mol. Pharmacol. 2008, 1, 255–269. [Google Scholar] [CrossRef]
  69. Samanta, A.; Hughes, T.E.T.; Moiseenkova-Bell, V.Y. Transient Receptor Potential (TRP) Channels. In Membrane Protein Complexes: Structure and Function; Harris, J.R., Boekema, E.J., Eds.; Springer: Singapore, 2018; pp. 141–165. [Google Scholar] [CrossRef]
  70. Zygmunt, P.M.; Högestätt, E.D. TRPA1. In Mammalian Transient Receptor Potential (TRP) Cation Channels: Volume I; Nilius, B., Flockerzi, V., Eds.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 583–630. [Google Scholar] [CrossRef]
  71. Cao, E.; Liao, M.; Cheng, Y.; Julius, D. TRPV1 structures in distinct conformations reveal activation mechanisms. Nature 2013, 504, 113–118. [Google Scholar] [CrossRef] [Green Version]
  72. Liao, M.; Cao, E.; Julius, D.; Cheng, Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature 2013, 504, 107–112. [Google Scholar] [CrossRef] [Green Version]
  73. Sappington, R.M.; Sidorova, T.; Long, D.J.; Calkins, D.J. TRPV1: Contribution to Retinal Ganglion Cell Apoptosis and Increased Intracellular Ca2+ with Exposure to Hydrostatic Pressure. Investig. Ophthalmol. Vis. Sci. 2009, 50, 717–728. [Google Scholar] [CrossRef] [Green Version]
  74. King, R.; Brain, S.D. Chapter 189—CGRP. In Handbook of Biologically Active Peptides, 2nd ed; Kastin, A.J., Ed.; Academic Press: Boston, MA, USA, 2013; pp. 1394–1401. [Google Scholar] [CrossRef]
  75. Russell, F.A.; King, R.; Smillie, S.-J.; Kodji, X.; Brain, S.D. Calcitonin Gene-Related Peptide: Physiology and Pathophysiology. Physiol. Rev. 2014, 94, 1099–1142. [Google Scholar] [CrossRef] [Green Version]
  76. Matteoli, M.; Haimann, C.; Torri-Tarelli, F.; Polak, J.M.; Ceccarelli, B.; De Camilli, P. Differential effect of alpha-latrotoxin on exocytosis from small synaptic vesicles and from large dense-core vesicles containing calcitonin gene-related peptide at the frog neuromuscular junction. Proc. Natl. Acad. Sci. USA 1988, 85, 7366–7370. [Google Scholar] [CrossRef] [PubMed]
  77. Fernandes, E.; Fernandes, M.; Keeble, J. The functions of TRPA1 and TRPV1: Moving away from sensory nerves. Br. J. Pharmacol. 2012, 166, 510–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Omari, S.A.; Adams, M.J.; Geraghty, D.P. Chapter Six—TRPV1 Channels in Immune Cells and Hematological Malignancies. In Advances in Pharmacology; Geraghty, D.P., Rash, L.D., Eds.; Academic Press: Cambridge, MA, USA, 2017; Volume 79, pp. 173–198. [Google Scholar]
  79. Naert, R.; López-Requena, A.; Talavera, K. TRPA1 Expression and Pathophysiology in Immune Cells. Int. J. Mol. Sci. 2021, 22, 11460. [Google Scholar] [CrossRef] [PubMed]
  80. Tamás, K.; Zsolt, B.; Erzsébet, L.; Enikő, T.P.; Nóra, E.; Ágnes, C.; Zoltán, P.; István, É.; Róbert, P.; Attila, T. Tissue-Specific Regulation of Microvascular Diameter: Opposite Functional Roles of Neuronal and Smooth Muscle Located Vanilloid Receptor-1. Mol. Pharmacol. 2008, 73, 1405. [Google Scholar] [CrossRef]
  81. Helley, M.P.; Abate, W.; Jackson, S.K.; Bennett, J.H.; Thompson, S.W.N. The expression of Toll-like receptor 4, 7 and co-receptors in neurochemical sub-populations of rat trigeminal ganglion sensory neurons. Neuroscience 2015, 310, 686–698. [Google Scholar] [CrossRef] [Green Version]
  82. Wadachi, R.; Hargreaves, K. Trigeminal nociceptors express TLR-4 and CD14: A mechanism for pain due to infection. J. Dent. Res. 2006, 85, 49–53. [Google Scholar] [CrossRef] [Green Version]
  83. Diogenes, A.; Ferraz, C.C.R.; Akopian, A.N.; Henry, M.A.; Hargreaves, K.M. LPS Sensitizes TRPV1 via Activation of TLR4 in Trigeminal Sensory Neurons. J. Dent. Res. 2011, 90, 759–764. [Google Scholar] [CrossRef]
  84. Meseguer, V.; Alpizar, Y.A.; Luis, E.; Tajada, S.; Denlinger, B.; Fajardo, O.; Manenschijn, J.-A.; Fernández-Peña, C.; Talavera, A.; Kichko, T.; et al. TRPA1 channels mediate acute neurogenic inflammation and pain produced by bacterial endotoxins. Nat. Commun. 2014, 5, 3125. [Google Scholar] [CrossRef] [Green Version]
  85. Ferraz, C.C.R.; Henry, M.A.; Hargreaves, K.M.; Diogenes, A. Lipopolysaccharide From Porphyromonas gingivalis Sensitizes Capsaicin-Sensitive Nociceptors. J. Endod. 2011, 37, 45–48. [Google Scholar] [CrossRef] [Green Version]
  86. Lagomarsino, V.N.; Kostic, A.D.; Chiu, I.M. Mechanisms of microbial–neuronal interactions in pain and nociception. Neurobiol. Pain 2021, 9, 100056. [Google Scholar] [CrossRef]
  87. Chiu, I.M.; Heesters, B.A.; Ghasemlou, N.; Von Hehn, C.A.; Zhao, F.; Tran, J.; Wainger, B.; Strominger, A.; Muralidharan, S.; Horswill, A.R.; et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature 2013, 501, 52–57. [Google Scholar] [CrossRef] [Green Version]
  88. Sun, X.; Yang, Z.; Nie, Y.; Hou, B. Microbial Communities in the Extraradicular and Intraradicular Infections Associated With Persistent Apical Periodontitis. Front. Cell. Infect. Microbiol. 2022, 11, 798367. [Google Scholar] [CrossRef]
  89. Eftekhari, S.; Salvatore, C.A.; Calamari, A.; Kane, S.A.; Tajti, J.; Edvinsson, L. Differential distribution of calcitonin gene-related peptide and its receptor components in the human trigeminal ganglion. Neuroscience 2010, 169, 683–696. [Google Scholar] [CrossRef] [Green Version]
  90. Messlinger, K.; Balcziak, L.K.; Russo, A.F. Cross-talk signaling in the trigeminal ganglion: Role of neuropeptides and other mediators. J. Neural Transm. 2020, 127, 431–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Messlinger, K. The big CGRP flood—Sources, sinks and signalling sites in the trigeminovascular system. J. Headache Pain 2018, 19, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Li, W.; Zhang, Z.; Li, X.; Cai, J.; Li, D.; Du, J.; Zhang, B.; Xiang, D.; Li, N.; Li, Y. CGRP derived from cardiac fibroblasts is an endogenous suppressor of cardiac fibrosis. Cardiovasc. Res. 2020, 116, 1335–1348. [Google Scholar] [CrossRef]
  93. Kim, Y.J.; Granstein, R.D. Roles of calcitonin gene-related peptide in the skin, and other physiological and pathophysiological functions. Brain Behav. Immun. Health 2021, 18, 100361. [Google Scholar] [CrossRef] [PubMed]
  94. Holzmann, B. Modulation of immune responses by the neuropeptide CGRP. Amino Acids 2013, 45, 1–7. [Google Scholar] [CrossRef] [PubMed]
  95. Yang, D.; Jacobson, A.; Meerschaert, K.A.; Sifakis, J.J.; Wu, M.; Chen, X.; Yang, T.; Zhou, Y.; Anekal, P.V.; Rucker, R.A.; et al. Nociceptor neurons direct goblet cells via a CGRP-RAMP1 axis to drive mucus production and gut barrier protection. Cell 2022, 185, 4190–4205.e4125. [Google Scholar] [CrossRef]
  96. Sert, S.; Sakallioğlu, U.; Lütfioğlu, M.; Aydoğdu, A.; Acarel, E.; Günaydın, M. Neurogenic inflammation in periimplant and periodontal disease: A case–control split-mouth study. Clin. Oral Implant. Res. 2019, 30, 800–807. [Google Scholar] [CrossRef] [PubMed]
  97. Lundy, F.T.; Shaw, C.; McKinnell, J.; Lamey, P.-J.; Linden, G.J. Calcitonin gene-related peptide in gingival crevicular fluid in periodontal health and disease. J. Clin. Periodontol. 1999, 26, 212–216. [Google Scholar] [CrossRef] [PubMed]
  98. Nelson, M.T.; Huang, Y.; Brayden, J.E.; Hescheler, J.; Standen, N.B. Arterial dilations in response to calcitonin gene-related peptide involve activation of K+ channels. Nature 1990, 344, 770–773. [Google Scholar] [CrossRef]
  99. Ma, W.; Dumont, Y.; Vercauteren, F.; Quirion, R. Lipopolysaccharide induces calcitonin gene-related peptide in the RAW264. 7 macrophage cell line. Immunology 2010, 130, 399–409. [Google Scholar] [CrossRef] [PubMed]
  100. Ma, W.; Quirion, R. Increased calcitonin gene-related peptide in neuroma and invading macrophages is involved in the up-regulation of interleukin-6 and thermal hyperalgesia in a rat model of mononeuropathy. J. Neurochem. 2006, 98, 180–192. [Google Scholar] [CrossRef] [PubMed]
  101. Sun, Y.; Tao, R.; Zhang, M.; Cao, X.; Wang, H.; Xue, L.; Wu, M. Expression of Calcitonin Gene-related Peptide in Rat Pulp and Periodontal Tissues by Indirect Immunofluorescence Method. Monoclon. Antibodies Immunodiagn. Immunother. 2013, 32, 404–408. [Google Scholar] [CrossRef] [Green Version]
  102. Niedermühlbichler, M.; Wiedermann, C.J. Suppression of superoxide release from human monocytes by somatostatin-related peptides. Regul. Pept. 1992, 41, 39–47. [Google Scholar] [CrossRef]
  103. Fox, F.E.; Kubin, M.; Cassin, M.; Niu, Z.; Hosoi, J.; Torii, H.; Granstein, R.D.; Trinchieri, G.; Rook, A.H. Calcitonin Gene-Related Peptide Inhibits Proliferation and Antigen Presentation by Human Peripheral Blood Mononuclear Cells: Effects on B7, Interleukin 10, and Interleukin12. J. Investig. Dermatol. 1997, 108, 43–48. [Google Scholar] [CrossRef] [Green Version]
  104. Takahashi, N.; Matsuda, Y.; Sato, K.; de Jong, P.R.; Bertin, S.; Tabeta, K.; Yamazaki, K. Neuronal TRPV1 activation regulates alveolar bone resorption by suppressing osteoclastogenesis via CGRP. Sci. Rep. 2016, 6, 29294. [Google Scholar] [CrossRef]
  105. Duan, Y.; Liang, Y.; Yang, F.; Ma, Y. Neural Regulations in Tooth Development and Tooth–Periodontium Complex Homeostasis: A Literature Review. Int. J. Mol. Sci. 2022, 23, 14150. [Google Scholar] [CrossRef]
  106. Johnson, M.B.; Young, A.D.; Marriott, I. The Therapeutic Potential of Targeting Substance P/NK-1R Interactions in Inflammatory CNS Disorders. Front. Cell. Neurosci. 2017, 10, 296. [Google Scholar] [CrossRef] [Green Version]
  107. Zieglgänsberger, W. Substance P and pain chronicity. Cell Tissue Res. 2019, 375, 227–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Mashaghi, A.; Marmalidou, A.; Tehrani, M.; Grace, P.M.; Pothoulakis, C.; Dana, R. Neuropeptide substance P and the immune response. Cell. Mol. Life Sci. 2016, 73, 4249–4264. [Google Scholar] [CrossRef] [Green Version]
  109. Graefe, S.B.; Mohiuddin, S.S. Biochemistry, Substance P.; StatPearls Publishing: St. Petersburg, FL, USA, 2022. [Google Scholar]
  110. Perner, C.; Flayer, C.H.; Zhu, X.; Aderhold, P.A.; Dewan, Z.N.A.; Voisin, T.; Camire, R.B.; Chow, O.A.; Chiu, I.M.; Sokol, C.L. Substance P Release by Sensory Neurons Triggers Dendritic Cell Migration and Initiates the Type-2 Immune Response to Allergens. Immunity 2020, 53, 1063–1077.e1067. [Google Scholar] [CrossRef] [PubMed]
  111. Pradeep, A.R.; Raj, S.; Aruna, G.; Chowdhry, S. Gingival crevicular fluid and plasma levels of neuropeptide Substance-P in periodontal health, disease and after nonsurgical therapy. J. Periodontal Res. 2009, 44, 232–237. [Google Scholar] [CrossRef] [PubMed]
  112. Avellán, N.-L.; Sorsa, T.; Tervahartiala, T.; Forster, C.; Kemppainen, P. Experimental tooth pain elevates substance P and matrix metalloproteinase-8 levels in human gingival crevice fluid. Acta Odontol. Scand. 2008, 66, 18–22. [Google Scholar] [CrossRef]
  113. Sacerdote, P.; Levrini, L. Peripheral Mechanisms of Dental Pain: The Role of Substance P. Mediat. Inflamm. 2012, 2012, 951920. [Google Scholar] [CrossRef]
  114. Niedermair, T.; Schirner, S.; Seebröker, R.; Straub, R.H.; Grässel, S. Substance P modulates bone remodeling properties of murine osteoblasts and osteoclasts. Sci. Rep. 2018, 8, 9199. [Google Scholar] [CrossRef] [Green Version]
  115. Yan, K.; Lin, Q.; Tang, K.; Liu, S.; Du, Y.; Yu, X.; Li, S. Substance P participates in periodontitis by upregulating HIF-1α and RANKL/OPG ratio. BMC Oral Health 2020, 20, 27. [Google Scholar] [CrossRef] [Green Version]
  116. Kim, S.; Dorscher-Kim, J. Effects of Substance P on Blood Flow of the Dental Pulp in Dogs. In Proceedings of the Substance P and Neurokinins; Springer-Verlag New York Inc.: New York, NY, USA, 1987; pp. 197–199. [Google Scholar]
  117. Augustyniak, D.; Roszkowiak, J.; Wiśniewska, I.; Skała, J.; Gorczyca, D.; Drulis-Kawa, Z. Neuropeptides SP and CGRP Diminish the Moraxella catarrhalis Outer Membrane Vesicle- (OMV-) Triggered Inflammatory Response of Human A549 Epithelial Cells and Neutrophils. Mediat. Inflamm. 2018, 2018, 4847205. [Google Scholar] [CrossRef] [Green Version]
  118. Iwasaki, M.a.A.Y.; Kaunitz, J.D. Recent advances in vasoactive intestinal peptide physiology and pathophysiology: Focus on the gastrointestinal system [version 1; peer review: 4 approved]. F1000Research 2019, 8, 1629. [Google Scholar] [CrossRef] [Green Version]
  119. Linden, G.J.; Mullally, B.H.; Burden, D.J.; Lamey, P.-J.; Shaw, C.; Ardill, J.; Lundy, F.T. Changes in vasoactive intestinal peptide in gingival crevicular fluid in response to periodontal treatment. J. Clin. Periodontol. 2002, 29, 484–489. [Google Scholar] [CrossRef] [PubMed]
  120. Luthman, J.; Friskopp, J.; Dahliöf, G.; Ahlström, U.; Sjöström, L.; Johansson, O. Immunohistochemical study of neurochemical markers in gingiva obtained from periodontitis-affected sites. J. Periodontal Res. 1989, 24, 267–278. [Google Scholar] [CrossRef] [PubMed]
  121. Abualsaud, N.; Caprio, L.; Galli, S.; Krawczyk, E.; Alamri, L.; Zhu, S.; Gallicano, G.I.; Kitlinska, J. Neuropeptide Y/Y5 Receptor Pathway Stimulates Neuroblastoma Cell Motility Through RhoA Activation. Front. Cell Dev. Biol. 2021, 8, 627090. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, Y.; Liu, C.-Y.; Chen, W.-C.; Shi, Y.-C.; Wang, C.-M.; Lin, S.; He, H.-F. Regulation of neuropeptide Y in body microenvironments and its potential application in therapies: A review. Cell Biosci. 2021, 11, 151. [Google Scholar] [CrossRef]
  123. Chen, W.-c.; Liu, Y.-b.; Liu, W.-f.; Zhou, Y.-y.; He, H.-f.; Lin, S. Neuropeptide Y Is an Immunomodulatory Factor: Direct and Indirect. Front. Immunol. 2020, 11, 580378. [Google Scholar] [CrossRef]
  124. Haririan, H.; Andrukhov, O.; Böttcher, M.; Pablik, E.; Wimmer, G.; Moritz, A.; Rausch-Fan, X. Salivary neuropeptides, stress, and periodontitis. J. Periodontol. 2018, 89, 9–18. [Google Scholar] [CrossRef] [PubMed]
  125. Winning, L.; El Karim, I.A.; Linden, G.J.; Irwin, C.R.; Killough, S.A.; Lundy, F.T. Differential regulation of NPY and SP receptor expression in STRO-1+ve PDLSCs by inflammatory cytokines. J. Periodontal Res. 2022, 57, 186–194. [Google Scholar] [CrossRef]
  126. Okragly, A.J.; Morin, S.M.; DeRosa, D.; Martin, A.P.; Johnson, K.W.; Johnson, M.P.; Benschop, R.J. Human mast cells release the migraine-inducing factor pituitary adenylate cyclase-activating polypeptide (PACAP). Cephalalgia 2018, 38, 1564–1574. [Google Scholar] [CrossRef]
  127. Maugeri, G.; D’Amico, A.G.; Musumeci, G.; Reglodi, D.; D’Agata, V. Effects of PACAP on Schwann Cells: Focus on Nerve Injury. Int. J. Mol. Sci. 2020, 21, 8233. [Google Scholar] [CrossRef]
  128. Kuri, B.A.; Chan, S.-A.; Smith, C.B. PACAP regulates immediate catecholamine release from adrenal chromaffin cells in an activity-dependent manner through a protein kinase C-dependent pathway. J. Neurochem. 2009, 110, 1214–1225. [Google Scholar] [CrossRef] [Green Version]
  129. Edvinsson, J.C.A.; Grell, A.-S.; Warfvinge, K.; Sheykhzade, M.; Edvinsson, L.; Haanes, K.A. Differences in pituitary adenylate cyclase-activating peptide and calcitonin gene-related peptide release in the trigeminovascular system. Cephalalgia 2020, 40, 1296–1309. [Google Scholar] [CrossRef] [PubMed]
  130. Delgado, M.; Munoz-Elias, E.J.; Gomariz, R.P.; Ganea, D. VIP and PACAP inhibit IL-12 production in LPS-stimulated macrophages. Subsequent effect on IFNγ synthesis by T cells. J. Neuroimmunol. 1999, 96, 167–181. [Google Scholar] [CrossRef]
  131. Trafton, J.A.; Abbadie, C.; Basbaum, A.I. Differential Contribution of Substance P and Neurokinin A to Spinal Cord Neurokinin-1 Receptor Signaling in the Rat. J. Neurosci. 2001, 21, 3656–3664. [Google Scholar] [CrossRef]
  132. Onaga, T. Tachykinin: Recent developments and novel roles in health and disease. Biomol. Concepts 2014, 5, 225–243. [Google Scholar] [CrossRef] [PubMed]
  133. Mishra, A.; Lal, G. Neurokinin receptors and their implications in various autoimmune diseases. Curr. Res. Immunol. 2021, 2, 66–78. [Google Scholar] [CrossRef] [PubMed]
  134. Lundy, F.T.; El Karim, I.A.; Linden, G.J. Neuropeptide Y (NPY) and NPY Y1 receptor in periodontal health and disease. Arch. Oral Biol. 2009, 54, 258–262. [Google Scholar] [CrossRef] [PubMed]
  135. Amara, S.G.; Jonas, V.; Rosenfeld, M.G.; Ong, E.S.; Evans, R.M. Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature 1982, 298, 240–244. [Google Scholar] [CrossRef]
  136. Fisher, L.A.; Kikkawa, D.O.; Rivier, J.E.; Amara, S.G.; Evans, R.M.; Rosenfeld, M.G.; Vale, W.W.; Brown, M.R. Stimulation of noradrenergic sympathetic outflow by calcitonin gene-related peptide. Nature 1983, 305, 534–536. [Google Scholar] [CrossRef]
  137. Brain, S.D.; Williams, T.J.; Tippins, J.R.; Morris, H.R.; MacIntyre, I. Calcitonin gene-related peptide is a potent vasodilator. Nature 1985, 313, 54–56. [Google Scholar] [CrossRef]
  138. Iyengar, S.; Ossipov, M.H.; Johnson, K.W. The role of calcitonin gene–related peptide in peripheral and central pain mechanisms including migraine. Pain 2017, 158, 543. [Google Scholar] [CrossRef] [Green Version]
  139. Hay, D.L.; Garelja, M.L.; Poyner, D.R.; Walker, C.S. Update on the pharmacology of calcitonin/CGRP family of peptides: IUPHAR Review 25. Br. J. Pharmacol. 2018, 175, 3–17. [Google Scholar] [CrossRef]
  140. v. Euler, U.S.; Gaddum, J.H. An unidentified depressor substance in certain tissue extracts. J. Physiol. 1931, 72, 74–87. [Google Scholar] [CrossRef]
  141. Krause, J.E.; Takeda, Y.; Hershey, A.D. Structure, Functions, and Mechanisms of Substance P Receptor Action. J. Investig. Dermatol. 1992, 98, S2–S7. [Google Scholar] [CrossRef] [Green Version]
  142. Arslan, H.; Yıldız, E.D.; Köseoğlu, S. Effects of endodontic treatment on salivary levels of CGRP and substance P: A pilot study. Restor. Dent. Endod. 2020, 45, e40. [Google Scholar] [CrossRef]
  143. Wakisaka, S. Neuropeptides in the dental pulp: Distribution, origins, and correlation. J. Endod. 1990, 16, 67–69. [Google Scholar] [CrossRef] [PubMed]
  144. Caviedes-Bucheli, J.; Muñoz, H.R.; Azuero-Holguín, M.M.; Ulate, E. Neuropeptides in Dental Pulp: The Silent Protagonists. J. Endod. 2008, 34, 773–788. [Google Scholar] [CrossRef]
  145. Kato, J.; Wakisaka, S.; Kurisu, K. Immunohistochemical Changes in the Distribution of Nerve Fibers in the Periodontal Ligament during an Experimental Tooth Movement of the Rat Molar. Acta Anat. 2008, 157, 53–62. [Google Scholar] [CrossRef]
  146. Caviedes-Bucheli, J.; Castellanos, F.; Vasquez, N.; Ulate, E.; Munoz, H.R. The influence of two reciprocating single-file and two rotary-file systems on the apical extrusion of debris and its biological relationship with symptomatic apical periodontitis. A systematic review and meta-analysis. Int. Endod. J. 2016, 49, 255–270. [Google Scholar] [CrossRef] [PubMed]
  147. Lundy, F.T.; Mullally, B.H.; Burden, D.J.; Lamey, P.-J.; Shaw, C.; Linden, G.J. Changes in substance P and neurokinin A in gingival crevicular fluid in response to periodontal treatment. J. Clin. Periodontol. 2000, 27, 526–530. [Google Scholar] [CrossRef] [PubMed]
  148. Burgener, B.; Ford, A.R.; Situ, H.; Fayad, M.I.; Hao, J.J.; Wenckus, C.S.; Johnson, B.R.; BeGole, E.A.; George, A. Biologic Markers for Odontogenic Periradicular Periodontitis. J. Endod. 2010, 36, 1307–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Stashenko, P.; Teles, R.; d’Souza, R. Periapical inflammatory responses and their modulation. Crit. Rev. Oral Biol. Med. 1998, 9, 498–521. [Google Scholar] [CrossRef] [PubMed]
  150. Takeda, K.; Akira, S. Toll-Like Receptors. Curr. Protoc. Immunol. 2007, 77, 14.12.11–14.12.13. [Google Scholar] [CrossRef] [PubMed]
  151. Lai, J.-P.; Ho, W.Z.; Kilpatrick, L.E.; Wang, X.; Tuluc, F.; Korchak, H.M.; Douglas, S.D. Full-length and truncated neurokinin-1 receptor expression and function during monocyte/macrophage differentiation. Proc. Natl. Acad. Sci. USA 2006, 103, 7771–7776. [Google Scholar] [CrossRef] [PubMed]
  152. Marriott, I.; Bost, K.L. IL-4 and IFN-γ Up-Regulate Substance P Receptor Expression in Murine Peritoneal Macrophages1. J. Immunol. 2000, 165, 182–191. [Google Scholar] [CrossRef]
  153. Simeonidis, S.; Castagliuolo, I.; Pan, A.; Liu, J.; Wang, C.-C.; Mykoniatis, A.; Pasha, A.; Valenick, L.; Sougioultzis, S.; Zhao, D.; et al. Regulation of the NK-1 receptor gene expression in human macrophage cells via an NF-κB site on its promoter. Proc. Natl. Acad. Sci. USA 2003, 100, 2957–2962. [Google Scholar] [CrossRef]
  154. Goldman, R.; Bar-Shavit, Z.; Shezen, E.; Terry, S.; Blumberg, S. Enhancement of Phagocytosis by Neurotensin, a Newly Found Biological Activity of the Neuropeptide. In Macrophages and Natural Killer Cells: Regulation and Function; Normann, S.J., Sorkin, E., Eds.; Springer US: Boston, MA, USA, 1982; pp. 133–141. [Google Scholar] [CrossRef]
  155. Hartung, H.-P. Activation of macrophages by neuropeptides. Brain Behav. Immun. 1988, 2, 275–281. [Google Scholar] [CrossRef]
  156. Ruff, M.R.; Wahl, S.M.; Pert, C.B. Substance P receptor-mediated chemotaxis of human monocytes. Peptides 1985, 6, 107–111. [Google Scholar] [CrossRef]
  157. Hartung, H.-P.; Toyka, K.V. Substance P, the Immune System and Inflammation. Int. Rev. Immunol. 1989, 4, 229–249. [Google Scholar] [CrossRef]
  158. Cozens, P.J.; Rowe, F.M. Substance-P is a potent inducer of tnf and IL-1 secretion by macrophages—A potential role for tnf in the pathogenesis of asthma. Immunobiology 1987, 175, 7. [Google Scholar]
  159. Saito, I.; Ishii, K.; Hanada, K.; Sato, O.; Maeda, T. Responses of calcitonin gene-related peptide-immunopositive nerve fibres in the periodontal ligament of rat molars to experimental tooth movement. Arch. Oral Biol. 1991, 36, 689–692. [Google Scholar] [CrossRef]
  160. Wakisaka, S.; Nishikawa, S.; Ichikawa, H.; Matsuo, S.; Takano, Y.; Akai, M. The distribution and origin of substance P-like immunoreactivity in the rat molar pulp and periodontal tissues. Arch. Oral Biol. 1985, 30, 813–818. [Google Scholar] [CrossRef]
  161. Heyeraas, K.J.; Kvinnsland, I.; Byers, M.R.; Jacobsen, E.B. Nerve fibers immunoreactive to protein gene product 9.5, calcitonin gene-related peptide, substance P, and neuropeptide Y in the dental pulp, periodontal ligament, and gingiva in cats. Acta Odontol. Scand. 1993, 51, 207–221. [Google Scholar] [CrossRef] [PubMed]
  162. Berggreen Jacobsen, E.; Fristad, I.; Heyeraas, K.J. Nerve fibers immunoreactive to calcitonin gene-related peptide, substance P, neuropeptide Y, and dopamine ß-hydroxylase in innervated and denervated oral tissues in ferrets. Acta Odontol. Scand. 1998, 56, 220–228. [Google Scholar] [CrossRef] [PubMed]
  163. Kimberly, C.L.; Byers, M.R. Inflammation of rat molar pulp and periodontium causes increased calcitonin gene-related peptide and axonal sprouting. Anat. Rec. 1988, 222, 289–300. [Google Scholar] [CrossRef]
  164. Khayat, B.G.; Byers, M.R.; Taylor, P.E.; Mecifi, K.; Kimberly, C.L. Responses of nerve fibers to pulpal inflammation and periapical lesions in rat molars demonstrated by calcitonin gene-related peptide immunocytochemistry. J. Endod. 1988, 14, 577–587. [Google Scholar] [CrossRef]
  165. Gašperšič, R.; Kovačič, U.; Cör, A.; Skalerič, U. Unilateral ligature-induced periodontitis influences the expression of neuropeptides in the ipsilateral and contralateral trigeminal ganglion in rats. Arch. Oral Biol. 2008, 53, 659–665. [Google Scholar] [CrossRef]
  166. Kabashima, H.; Nagata, K.; Maeda, K.; Iijima, T. Involvement of substance P, mast cells, TNF-α and ICAM-1 in the infiltration of inflammatory cells in human periapical granulomas. J. Oral Pathol. Med. 2002, 31, 175–180. [Google Scholar] [CrossRef]
  167. de Avila, E.D.; de Molon, R.S.; de Godoi Gonçalves, D.A.; Camparis, C.M. Relationship between levels of neuropeptide Substance P in periodontal disease and chronic pain: A literature review. J. Investig. Clin. Dent. 2014, 5, 91–97. [Google Scholar] [CrossRef]
  168. Ferrin, J.; Kirakodu, S.; Jensen, D.; Al-Attar, A.; Peyyala, R.; Novak, M.J.; Dawson, D.; Al-Sabbagh, M.; Stromberg, A.J.; Orraca, L.; et al. Gene expression analysis of neuropeptides in oral mucosa during periodontal disease in non-human primates. J. Periodontol. 2018, 89, 858–866. [Google Scholar] [CrossRef] [PubMed]
  169. Liinden, G.J.; McKinneil, J.; Shaw, C.; Lundy, F.T. Substance P and neurokinin A in gingival crevicular fluid in periodontal health and disease. J. Clin. Periodontol. 1997, 24, 799–803. [Google Scholar] [CrossRef]
  170. Hanioka, T.; Takaya, K.; Matsumori, Y.; Matsuse, R.; Shizukuishi, S. Relationship of the substance P to indicators of host response in human gingival crevicular fluid. J. Clin. Periodontol. 2000, 27, 262–266. [Google Scholar] [CrossRef] [PubMed]
  171. Lundy, F.T.; Salmon, A.L.; Lamey, P.-J.; Shaw, C.; Linden, G.J. Carboxypeptidase-mediated metabolism of calcitonin gene-related peptide in human gingival crevicular fluid—A rôle in periodontal inflammation? J. Clin. Periodontol. 2000, 27, 499–505. [Google Scholar] [CrossRef]
  172. Owan, I.; Ibaraki, K. The role of calcitonin gene-related peptide (CGRP) in macrophages: The presence of functional receptors and effects on proliferation and differentiation into osteoclast-like cells. Bone Miner. 1994, 24, 151–164. [Google Scholar] [CrossRef]
  173. Fernandez, S.; Knopf, M.A.; Bjork, S.K.; McGillis, J.P. Bone Marrow-Derived Macrophages Express Functional CGRP Receptors and Respond to CGRP by Increasing Transcription of c-fos and IL-6 mRNA. Cell. Immunol. 2001, 209, 140–148. [Google Scholar] [CrossRef] [PubMed]
  174. Frielingsdorf, H.; Simpson, D.R.; Thal, L.J.; Pizzo, D.P. Nerve growth factor promotes survival of new neurons in the adult hippocampus. Neurobiol. Dis. 2007, 26, 47–55. [Google Scholar] [CrossRef] [PubMed]
  175. Ulupinar, E.; Jacquin, M.F.; Erzurumlu, R.S. Differential effects of NGF and NT-3 on embryonic trigeminal axon growth patterns. J. Comp. Neurol. 2000, 425, 202–218. [Google Scholar] [CrossRef]
  176. Minnone, G.; De Benedetti, F.; Bracci-Laudiero, L. NGF and Its Receptors in the Regulation of Inflammatory Response. Int. J. Mol. Sci. 2017, 18, 1028. [Google Scholar] [CrossRef] [Green Version]
  177. Salio, C.; Averill, S.; Priestley, J.V.; Merighi, A. Costorage of BDNF and neuropeptides within individual dense-core vesicles in central and peripheral neurons. Dev. Neurobiol. 2007, 67, 326–338. [Google Scholar] [CrossRef]
  178. Ehrhard, P.B.; Ganter, U.; Stalder, A.; Bauer, J.; Otten, U. Expression of functional trk protooncogene in human monocytes. Proc. Natl. Acad. Sci. USA 1993, 90, 5423–5427. [Google Scholar] [CrossRef]
  179. Kurihara, H.; Shinohara, H.; Yoshino, H.; Takeda, K.; Shiba, H. Neurotrophins in Cultured Cells from Periodontal Tissues. J. Periodontol. 2003, 74, 76–84. [Google Scholar] [CrossRef]
  180. Ochi, K.; Saito, I.; Hanada, K.; Maeda, T. Expression of TrkB-like immunoreactivity in non-neural cells of rat periodontal ligament. Arch. Oral Biol. 1997, 42, 455–464. [Google Scholar] [CrossRef] [PubMed]
  181. Caroleo, M.C.; Costa, N.; Bracci-Laudiero, L.; Aloe, L. Human monocyte/macrophages activate by exposure to LPS overexpress NGF and NGF receptors. J. Neuroimmunol. 2001, 113, 193–201. [Google Scholar] [CrossRef] [PubMed]
  182. la Sala, A.; Corinti, S.; Federici, M.; Saragovi, H.U.; Girolomoni, G. Ligand activation of nerve growth factor receptor TrkA protects monocytes from apoptosis. J. Leukoc. Biol. 2000, 68, 104–110. [Google Scholar] [CrossRef] [PubMed]
  183. Bracci-Laudiero, L.; Aloe, L.; Caroleo, M.C.; Buanne, P.; Costa, N.; Starace, G.; Lundeberg, T. Endogenous NGF regulates CGRP expression in human monocytes, and affects HLA-DR and CD86 expression and IL-10 production. Blood 2005, 106, 3507–3514. [Google Scholar] [CrossRef] [Green Version]
  184. Williams, K.S.; Killebrew, D.A.; Clary, G.P.; Seawell, J.A.; Meeker, R.B. Differential regulation of macrophage phenotype by mature and pro-nerve growth factor. J. Neuroimmunol. 2015, 285, 76–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Sun, J.; Ramnath, R.D.; Zhi, L.; Tamizhselvi, R.; Bhatia, M. Substance P enhances NF-κB transactivation and chemokine response in murine macrophages via ERK1/2 and p38 MAPK signaling pathways. Am. J. Physiol. Cell Physiol. 2008, 294, C1586–C1596. [Google Scholar] [CrossRef]
  186. Lee, S.-K.; Pi, S.-H.; Kim, S.-H.; Min, K.-S.; Lee, H.-J.; Chang, H.-S.; Kang, K.-H.; Kim, H.-R.; Shin, H.-I.; Kim, E.-C. Substance P regulates macrophage inflammatory protein 3α/chemokine C-C ligand 20 (CCL20) with heme oxygenase-1 in human periodontal ligament cells. Clin. Exp. Immunol. 2007, 150, 567–575. [Google Scholar] [CrossRef]
  187. Keane, M.P.; Strieter, R.M.; Belperio, J.A. Macrophage inflammatory protein. In Encyclopedia of Respiratory Medicine; Laurent, G.J., Shapiro, S.D., Eds.; Academic Press: Oxford, UK, 2006; pp. 1–5. [Google Scholar] [CrossRef]
  188. Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte Chemoattractant Protein-1 (MCP-1): An Overview. J. Interferon Cytokine Res. 2009, 29, 313–326. [Google Scholar] [CrossRef]
  189. Jiang, M.H.; Chung, E.; Chi, G.F.; Ahn, W.; Lim, J.E.; Hong, H.S.; Kim, D.W.; Choi, H.; Kim, J.; Son, Y. Substance P induces M2-type macrophages after spinal cord injury. Neuroreport 2012, 23, 786–792. [Google Scholar] [CrossRef]
  190. Hong, H.S.; Son, Y. Substance P ameliorates collagen II-induced arthritis in mice via suppression of the inflammatory response. Biochem. Biophys. Res. Commun. 2014, 453, 179–184. [Google Scholar] [CrossRef]
  191. Montana, G.; Lampiasi, N. Substance P Induces HO-1 Expression in RAW 264.7 Cells Promoting Switch towards M2-Like Macrophages. PLoS ONE 2016, 11, e0167420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Zhang, M.; Nakamura, K.; Kageyama, S.; Lawal, A.O.; Gong, K.W.; Bhetraratana, M.; Fujii, T.; Sulaiman, D.; Hirao, H.; Bolisetty, S.; et al. Myeloid HO-1 modulates macrophage polarization and protects against ischemia-reperfusion injury. JCI Insight 2019, 3, e120596. [Google Scholar] [CrossRef] [PubMed]
  193. Lim, J.E.; Chung, E.; Son, Y. A neuropeptide, Substance-P, directly induces tissue-repairing M2 like macrophages by activating the PI3K/Akt/mTOR pathway even in the presence of IFNγ. Sci. Rep. 2017, 7, 9417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Pinho-Ribeiro, F.A.; Verri, W.A.; Chiu, I.M. Nociceptor Sensory Neuron–Immune Interactions in Pain and Inflammation. Trends Immunol. 2017, 38, 5–19. [Google Scholar] [CrossRef] [Green Version]
  195. Rechenberg, D.K.; Munir, A.; Zehnder, M. Correlation between the clinically diagnosed inflammatory process and periapical index scores in severely painful endodontically involved teeth. Int. Endod. J. 2021, 54, 172–180. [Google Scholar] [CrossRef] [PubMed]
  196. Wideman, T.H.; Edwards, R.R.; Walton, D.M.; Martel, M.O.; Hudon, A.; Seminowicz, D.A. The Multimodal Assessment Model of Pain: A Novel Framework for Further Integrating the Subjective Pain Experience Within Research and Practice. Clin. J. Pain 2019, 35, 212. [Google Scholar] [CrossRef]
  197. Wilcox, C.E.; Mayer, A.R.; Teshiba, T.M.; Ling, J.; Smith, B.W.; Wilcox, G.L.; Mullins, P.G. The Subjective Experience of Pain: An FMRI Study of Percept-Related Models and Functional Connectivity. Pain Med. 2015, 16, 2121–2133. [Google Scholar] [CrossRef] [Green Version]
  198. Tarnawski, L.; Olofsson, P.S. Inflammation neuroscience: Neuro-immune crosstalk and interfaces. Clin. Transl. Immunol. 2021, 10, e1352. [Google Scholar] [CrossRef]
  199. Takeda, M.; Matsumoto, S.; Sessle, B.J.; Shinoda, M.; Iwata, K. Peripheral and Central Mechanisms of Trigeminal Neuropathic and Inflammatory Pain. J. Oral Biosci. 2011, 53, 318–329. [Google Scholar] [CrossRef]
  200. Khan, A.A.; Owatz, C.B.; Schindler, W.G.; Schwartz, S.A.; Keiser, K.; Hargreaves, K.M. Measurement of Mechanical Allodynia and Local Anesthetic Efficacy in Patients with Irreversible Pulpitis and Acute Periradicular Periodontitis. J. Endod. 2007, 33, 796–799. [Google Scholar] [CrossRef]
  201. Owatz, C.B.; Khan, A.A.; Schindler, W.G.; Schwartz, S.A.; Keiser, K.; Hargreaves, K.M. The Incidence of Mechanical Allodynia in Patients with Irreversible Pulpitis. J. Endod. 2007, 33, 552–556. [Google Scholar] [CrossRef] [PubMed]
  202. Schou, W.S.; Ashina, S.; Amin, F.M.; Goadsby, P.J.; Ashina, M. Calcitonin gene-related peptide and pain: A systematic review. J. Headache Pain 2017, 18, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Shi, X.; Wang, L.; Li, X.; Sahbaie, P.; Kingery, W.S.; Clark, J.D. Neuropeptides Contribute to Peripheral Nociceptive Sensitization by Regulating Interleukin-1β Production in Keratinocytes. Anesth. Analg. 2011, 113, 175. [Google Scholar] [CrossRef] [Green Version]
  204. Sun, R.-Q.; Lawand, N.B.; Willis, W.D. The role of calcitonin gene-related peptide (CGRP) in the generation and maintenance of mechanical allodynia and hyperalgesia in rats after intradermal injection of capsaicin. PAIN 2003, 104, 201–208. [Google Scholar] [CrossRef]
  205. França, G.M.D.; Carmo, A.F.d.; Costa Neto, H.; Andrade, A.L.D.L.d.; Lima, K.C.d.; Galvão, H.C. Macrophages subpopulations in chronic periapical lesions according to clinical and morphological aspects. Braz. Oral Res. 2019, 33, e047. [Google Scholar] [CrossRef] [Green Version]
  206. Toriya, Y.; Hashiguchi, I.; Maeda, K. Immunohistochemical examination of the distribution of macrophages and CGRP-immunoreactive nerve fibers in induced rat periapical lesions. Dent. Traumatol. 1997, 13, 6–12. [Google Scholar] [CrossRef] [PubMed]
  207. Pereira Faustino, I.S.; Azevedo, R.S.; Takahama, A. Metalloproteinases 2 and 9 Immunoexpression in Periapical Lesions from Primary Endodontic Infection: Possible Relationship with the Histopathological Diagnosis and the Presence of Pain. J. Endod. 2016, 42, 547–551. [Google Scholar] [CrossRef]
  208. Hong, C.-Y.; Lin, S.-K.; Kok, S.-H.; Cheng, S.-J.; Lee, M.-S.; Wang, T.-M.; Chen, C.-S.; Lin, L.-D.; Wang, J.-S. The role of lipopolysaccharide in infectious bone resorption of periapical lesion. J. Oral Pathol. Med. 2004, 33, 162–169. [Google Scholar] [CrossRef]
  209. Lin, S.-K.; Kok, S.-H.; Kuo, M.Y.-P.; Wang, T.-J.; Wang, J.-T.; Yeh, F.T.-C.; Hsiao, M.; Lan, W.-H.; Hong, C.-Y. Sequential expressions of MMP-1, TIMP-1, IL-6, and COX-2 genes in induced periapical lesions in rats. Eur. J. Oral Sci. 2002, 110, 246–253. [Google Scholar] [CrossRef]
  210. Jain, A.; Bahuguna, R. Role of matrix metalloproteinases in dental caries, pulp and periapical inflammation: An overview. J. Oral Biol. Craniofacial Res. 2015, 5, 212–218. [Google Scholar] [CrossRef] [Green Version]
  211. Barreiros, D.; Nelson Filho, P.; Paula-Silva, F.W.G.; Oliveira, K.M.H.d.; Lucisano, M.P.; Rossi, A.D.; Silva, L.A.B.; Küchler, E.C.; Silva, R.A.B. MMP2 and MMP9 are Associated with Apical Periodontitis Progression and Might be Modulated by TLR2 and MyD88. Braz. Dent. J. 2018, 29, 43–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Luchian, I.; Goriuc, A.; Sandu, D.; Covasa, M. The Role of Matrix Metalloproteinases (MMP-8, MMP-9, MMP-13) in Periodontal and Peri-Implant Pathological Processes. Int. J. Mol. Sci. 2022, 23, 1806. [Google Scholar] [CrossRef] [PubMed]
  213. Laronha, H.; Caldeira, J. Structure and Function of Human Matrix Metalloproteinases. Cells 2020, 9, 1076. [Google Scholar] [CrossRef]
  214. Elgezawi, M.; Haridy, R.; Almas, K.; Abdalla, M.A.; Omar, O.; Abuohashish, H.; Elembaby, A.; Christine Wölfle, U.; Siddiqui, Y.; Kaisarly, D. Matrix Metalloproteinases in Dental and Periodontal Tissues and Their Current Inhibitors: Developmental, Degradational and Pathological Aspects. Int. J. Mol. Sci. 2022, 23, 8929. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nociceptor neurons express receptor molecules that recognize PAMPs, DAMPs, cytokines, and neuropeptides. This results in the activation of downstream signaling pathways that can contribute to neurogenic inflammation. Initialisms are listed in Appendix A.
Figure 1. Nociceptor neurons express receptor molecules that recognize PAMPs, DAMPs, cytokines, and neuropeptides. This results in the activation of downstream signaling pathways that can contribute to neurogenic inflammation. Initialisms are listed in Appendix A.
Biomolecules 13 01193 g001
Figure 2. In addition to releasing neuropeptides, macrophages have been shown to express neuropeptide-specific receptors, indicating a significant role in neuro-immune cross-talk. Initialisms are listed in Appendix A.
Figure 2. In addition to releasing neuropeptides, macrophages have been shown to express neuropeptide-specific receptors, indicating a significant role in neuro-immune cross-talk. Initialisms are listed in Appendix A.
Biomolecules 13 01193 g002
Figure 3. Bi-directional cross-talk between nociceptor neurons and macrophages has been implicated in inflammation, resulting in pain and bone resorption in apical periodontitis. Initialisms are listed in Appendix A.
Figure 3. Bi-directional cross-talk between nociceptor neurons and macrophages has been implicated in inflammation, resulting in pain and bone resorption in apical periodontitis. Initialisms are listed in Appendix A.
Biomolecules 13 01193 g003
Table 1. Neuropeptides (NPs) identified in apical periodontitis (AP), cells known to release NPs, cells shown to express receptors for NPs, and known functional effects of NPs in AP.
Table 1. Neuropeptides (NPs) identified in apical periodontitis (AP), cells known to release NPs, cells shown to express receptors for NPs, and known functional effects of NPs in AP.
Neuropeptides in APCells Releasing NPsCells Expressing NP ReceptorsFunctional Effects in AP
Calcitonin Gene-Related Peptide
(CGRP)
Sensory neurons [9,75]
T cells [91]
Monocytes [91]
Endothelial cells [75]
Fibroblasts [92,93]
Adipocytes [75]
Sensory neurons [90]
Arterial vessels, Mononuclear immune cells, Schwann cells [91]
Th1 cells [94]
Langerhans cells [94]
Monocytes [94]
Macrophages [94]
Dendritic cells [94]
CD34+ hematopoietic progenitor cells [94]
B & T lymphocytes [94]
Epithelial cells [95]
Extensive degradation, resulting in reduced levels of CGRP observed in the GCF of diseased periodontal tissues [96,97]
Vasodilation [98]
Activation of innate and adaptive immune cells [99,100]
Increase in blood flow [101]
Prevention of monocyte/macrophage activation [102,103]
Stimulation of osteogenesis [104,105]
Endothelial cell proliferation [101]
Substance P
(SP)
Neurons [106,107]
Astrocytes [108]
Microglia [107,108]
Epithelial cells [108]
Endothelial cells [108]
T cells [108]
Macrophages [108]
Dendritic cells [106,108]
Eosinophils [108]
Mesenchymal stem cells [108]
Leukocytes [106]
Natural killer cells [106]
Mast cells [106]
Neutrophils [106]
Epithelial cells [108]
Endothelial cells [106,108,109]
White blood cells [109]
Fibroblasts [108,109]
Neurons [90,108,109]
Smooth muscle cells [106,108]
B & T lymphocytes [106,108]
Natural killer cells [108]
Dendritic cells [108,110]
Monocytes [106,108]
Macrophages [106,108]
Microglia [106,108]
Astrocytes [106,108]
Eosinophils [106,108]
Mast cells [106,108]
Neutrophils [106]
Increased levels observed in GCF of diseased periodontal tissues [96,111]
Desensitization of nociceptors [108]
Neuronal activation [112]
Onset of pain and inflammation [113]
Inhibition of osteoblast differentiation [114]
Induction of osteoclastogenesis [115]
Increase in blood flow [113,116]
Reduction in pro-inflammatory response of neutrophil, epithelial cells [117]
Vasoactive Intestinal Polypeptide
(VIP)
Neurons [118]
T cells [118]
B cells [118]
Mast cells [118]
Eosinophils [118]
Epithelial cells [118]
T cells [118]
Macrophages [118]
Dendritic cells [118]
Mast cells [118]
Neutrophils [118]
Smooth muscle cells [118]
Endothelial cells [118]
Pancreatic B cells [118]
Found in increased levels in GCF of periodontitis patients; reduced levels as inflammation reduced [119]
VIP reactivity observed in gingival biopsy, but no discernible difference compared to clinically healthy sites [120]
Promotion of bone regeneration [105]
Neuropeptide Y
(NPY)
Neurons [121]
Pancreatic β cells [122]
Vascular smooth muscle cells [123]
Monocytes [123]
Macrophages [123]
Enterocytes [123]
Endothelial cells [121]
Langerhans cells [123]
Microglia [123]
B & T lymphocytes [123]
Neurons [121]
Monocytes [123]
Macrophages [122,123]
Lymphocytes [123]
Dendritic Cells [123]
Neutrophils [123]
B & T Lymphocytes [123]
Leukocytes [123]
Microglia [123]
Osteoblasts [122]
Endothelial Cells [122]
PDL Stem Cells [105]
Proposed to be involved in initiation and regulation of periodontitis [105]
Reduced levels observed in diseased sites of periodontitis patients [96]
High levels observed in the saliva of periodontitis patients [124]
Promotion of osteogenesis [105]
Increase in PDL stem cells’ osteogenic capacity in vitro [125]
Pituitary Adenylate Cyclase-Activating Peptide
(PACAP)
Mast cells [126]
Schwann cells [127]
Pancreatic cells [128]
Sensory nerve terminals [128,129]
Pituitary cells [127]
Schwann cells [127]
Satellite glial cells [129]
Hepatocytes [129]
Osteoblasts [129]
Promotion of bone regeneration [105]
Dose-dependent inhibition of IL-12 in LPS-activated macrophages [130]
Neurokinin A
(NKA)
Sensory neurons [131]Sensory neurons [132]
B & T cells [133]
Dendritic cells [133]
Macrophages [133]
Co-released with SP [131]
Increased levels observed in the GCF of diseased periodontal tissues [134]
Decreased levels of NK-A observed in diseased periodontal tissue [96]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Menon, N.; Kishen, A. Nociceptor–Macrophage Interactions in Apical Periodontitis: How Biomolecules Link Inflammation with Pain. Biomolecules 2023, 13, 1193. https://doi.org/10.3390/biom13081193

AMA Style

Menon N, Kishen A. Nociceptor–Macrophage Interactions in Apical Periodontitis: How Biomolecules Link Inflammation with Pain. Biomolecules. 2023; 13(8):1193. https://doi.org/10.3390/biom13081193

Chicago/Turabian Style

Menon, Nandita, and Anil Kishen. 2023. "Nociceptor–Macrophage Interactions in Apical Periodontitis: How Biomolecules Link Inflammation with Pain" Biomolecules 13, no. 8: 1193. https://doi.org/10.3390/biom13081193

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

Article Metrics

Back to TopTop