Next Article in Journal
Correction: Moreno-Fonseca et al. Postfire Scenarios Shape Dung Beetle Communities in the Orinoquía Riparian Forest–Savannah Transition. Biology 2025, 14, 423
Previous Article in Journal
Machine Learning Analysis of Maize Seedling Traits Under Drought Stress
Previous Article in Special Issue
The Neural Mechanisms of Visual and Vestibular Interaction in Self-Motion Perception
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Injury-Driven Structural and Molecular Modifications in Nociceptors

by
Mario García-Domínguez
1,2,3
1
Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain
2
Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
3
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
Biology 2025, 14(7), 788; https://doi.org/10.3390/biology14070788
Submission received: 7 June 2025 / Revised: 27 June 2025 / Accepted: 27 June 2025 / Published: 29 June 2025
(This article belongs to the Special Issue Mechanisms Underlying Neuronal Network Activity)

Simple Summary

Following tissue injury, nociceptors (the primary sensory neurons responsible for detecting pain) undergo extensive structural and functional modifications. These changes are pivotal in the development and maintenance of both acute and chronic pain. Structurally, injury leads to alterations in the morphology and synaptic connectivity of nociceptors, which influence their excitability and patterns of neural communication. Simultaneously, molecular adaptations occur, such as changes in the expression and function of ion channels, receptors, and intracellular signaling pathways, as well as shifts in gene transcription that modulate nociceptive signal processing. This review consolidates current insights into the mechanisms underlying nociceptor plasticity after injury.

Abstract

Peripheral tissue injury initiates a multifaceted cascade of structural and molecular modifications within nociceptors, the primary sensory neurons tasked with detecting noxious stimuli. These alterations play a crucial role in the induction and maintenance of pain states, encompassing acute and chronic pain. Structural remodeling includes alterations in axonal architecture, dendritic morphology, and synaptic connectivity, collectively impacting nociceptor excitability and functional integration. Simultaneously, molecular adaptations comprise the regulation of ion channels, receptor expression, and intracellular signaling pathways, as well as transcriptional reprogramming that modulates nociceptive signaling. This review synthesizes current evidence regarding the cellular and molecular bases of injury-induced plasticity in nociceptors, identifying prospective targets for therapeutic intervention to counteract maladaptive sensitization. Elucidating these processes is critical for the advancement of pain treatment strategies and for enhancing clinical outcomes in individuals experiencing neuropathic pain secondary to tissue injury.

1. Introduction

The International Association for the Study of Pain (IASP) describes pain as “an unpleasant sensory and emotional experience often associated with actual or potential tissue damage” [1,2]. This definition underscores the complex nature of pain, encompassing both physical and emotional components, and usually reflecting underlying health conditions that require clinical intervention [3,4,5]. Pain perception is initiated by the activation of nociceptors, specialized sensory neurons localized within peripheral tissues such as the skin, musculature, and internal organs [6]. Nociceptors, specialized in detecting noxious stimuli such as mechanical pressure, extreme thermal conditions, and chemical alterations indicative of potential tissue injury [7], express many transduction molecules and ion channels that mediate the conversion of these stimuli into electrical signals [8]. These signals are transmitted centrally to the spinal cord and brain, where they are processed and interpreted as the subjective experience of pain. Importantly, nociceptors exhibit considerable heterogeneity in both structure and function, comprising distinct subpopulations characterized by variations in activation thresholds, neurochemical profiles, conduction velocities, and patterns of target tissue innervation [9]. This diversity underlies the complexity and specificity of nociceptive signaling.
Recent evidence challenges the traditional view of nociceptors as passive transmitters of noxious stimuli, revealing their plasticity and active involvement in the modulation of pain signaling under both physiological and pathological conditions [10]. Nociceptors are dynamic, responsive cells capable of adapting to changing environmental stimuli, especially in the context of injury, inflammation, or persistent pathological conditions [11]. In response to peripheral tissue injury, nociceptors undergo several molecular and structural changes that collectively rewire their sensory capacity. At the transcriptional level, there is a strong expression of genes encoding voltage-gated sodium channels (VGSCs), notably Nav1.7, Nav1.8, and Nav1.9, which contribute to increased neuronal excitability by reducing the threshold required for the action potential initiation [12,13,14]. Concurrently, voltage-gated calcium channels (VGCCs), such as Cav2.2, become more abundant or more readily trafficked to the membrane, facilitating enhanced neurotransmitter release at central terminals [15]. Transient receptor potential (TRP) channels, such as TRPV1 and TRPA1, are sensitized through post-translational modifications, including phosphorylation by protein kinases (such as PKC, PKA, and MAPKs), which are triggered downstream of several pro-inflammatory mediators including prostaglandins, bradykinin, and nerve growth factor (NGF) [16]. This leads to increased neuronal excitability and reduced activation thresholds, enabling innocuous stimuli to induce pain (commonly termed allodynia) and potentiating the response to noxious stimuli, a phenomenon referred to as hyperalgesia [17].
Moreover, nociceptors mobilize receptors for pro-inflammatory cytokines and chemokines, including TNF-α, IL-1β, and CCL2, which further potentiate intracellular signaling cascades through activation of NF-κB and other transcription factors [18,19,20]. These pathways not only sustain peripheral sensitization but also influence some epigenetic regulators, promoting long-lasting changes in gene expression [21,22]. Axonal transport and translation of mRNAs encoding ion channels and signaling molecules within nociceptive terminals constitute a key mechanism for activity-dependent modulation of nociceptive signaling [23,24]. Furthermore, nociceptors increase the synthesis and release of neuropeptides such as substance P and calcitonin gene-related peptide (CGRP), which act both peripherally to promote vasodilation and immune cell recruitment, and centrally to enhance excitatory neurotransmission in the spinal dorsal horn [25,26].
Novel methodological innovations (like single-cell transcriptomics, in vivo calcium imaging, chemogenetics, and optogenetics) have deepened our ability to dissect nociceptor subtypes and their activity patterns with unprecedented resolution [27,28,29,30]. All of these technologies have identified injury-induced gene expression profiles that extend beyond typical stress responses, involving the activation of developmental and regenerative transcriptional pathways [31].
Collectively, these findings underscore the pivotal role of nociceptor molecular plasticity in driving the transition from acute nociceptive responses to persistent chronic pain states. The dynamic regulation of ion channel expression, receptor sensitization, intracellular signaling pathways, and neuropeptide release not only facilitates neuronal excitability but also contributes to long-term alterations in pain processing circuits. Understanding these adaptations provides valuable insight into the pathophysiology of chronic pain and identifies numerous potential molecular targets that could be exploited for the development of novel analgesic therapies aimed at preventing or reversing maladaptive nociceptive sensitization.

2. Structural and Functional Aspects of Nociceptors

Nociceptors constitute a specialized subset of primary afferent neurons responsible for the detection, transduction, and propagation of noxious stimuli, which are defined as stimuli capable of inducing actual or potential tissue damage (Figure 1) [32]. As the primary sensory receptors for pain, nociceptors play a pivotal role in enabling the organism to detect and appropriately respond to harmful exogenous and endogenous signals [33]. The cell bodies of these neurons reside in peripheral sensory ganglia, mainly within the dorsal root ganglia (DRG) and trigeminal ganglia (TG) [34]. Each nociceptor projects a bifurcating axon, with one branch projecting peripherally to innervate cutaneous, musculoskeletal, and visceral structures, and the other projecting centrally to establish synaptic connections within defined laminar regions of the dorsal horn of the spinal cord [35].
Nociceptors exhibit considerable heterogeneity with respect to their conduction velocity, degree of myelination, and stimulus modality preference. Aδ fibers, characterized by thin myelination and intermediate conduction velocities (5–35 m/s), are typically associated with the rapid transmission of sharp, localized pain [36]. C fibers, which are unmyelinated and conduct more slowly (0.5–2 m/s), are involved in the mediation of dull, burning, and poorly localized pain sensations [37]. This physiological dichotomy corresponds to a molecular diversification of transducer proteins and ion channels, which confer polymodal or unimodal sensitivities to nociceptors [38]. Polymodal nociceptors are responsive to many noxious stimuli, while unimodal nociceptors are selectively sensitive to particular types of stimuli [39]. These functional phenotypes are regulated through developmental and activity-dependent processes and are influenced by the tissue environment [40]. Nociceptor transduction of injurious stimuli (mechanical, thermal, and chemical) is mediated by a complex array of ion channels and receptor proteins strategically localized at the peripheral terminal membranes, which convert many noxious modalities into electrical signals [8].
The physiological and molecular diversity of nociceptors underlies their varied functional roles in pain detection. This heterogeneity arises from the differential expression of specialized transducer proteins and ion channels, which facilitate the detection and transduction of a wide spectrum of noxious stimuli via distinct mechanisms [41]. TRP channels function as polymodal sensors essential for nociception [42]. TRPV1 responds to noxious heat (>42 °C), acidic pH, and capsaicin, allowing cation influx that depolarizes nociceptors and activates Ca2+-dependent pathways (PKC and CaMK), thereby sensitizing the channel and promoting hyperalgesia [43]. TRPA1 detects electrophilic irritants and inflammatory mediators, resulting in cation influx, nociceptor depolarization, and subsequent activation of PLC and MAPK signaling pathways, which enhance neurogenic inflammation and mechanical hypersensitivity [44]. TRPM8 is activated by cool temperatures and menthol, and modulates pain pathways, generally counteracting TRPV1-mediated heat pain [45]. Mechanical nociception is mediated mainly by Piezo1 and Piezo2 channels, which open in response to membrane stretch or shear forces, permitting rapid cation influx and membrane depolarization [46]. Piezo2 is abundantly expressed in DRG neurons and mediates the detection of light touch and mechanical nociception [47], whereas Piezo1 plays a contributory role under conditions of tissue stress [48]. Additional nociceptive inputs arise from acid-sensing ion channels (ASICs), which detect extracellular acidification by allowing Na+ and Ca2+ influx [49], and purinergic P2X receptors (notably P2X3), which become activated by extracellular ATP released from damaged cells, thereby facilitating the sustained transmission of chronic pain signals [50].
The combined ionic influx (principally Na+ and Ca2+) from these channels generates receptor potentials that activate VGSCs (Nav1.7, Nav1.8, and Nav1.9), amplifying depolarization into action potentials [51]. Nav1.7 controls action potential threshold [52], while Nav1.8 and Nav1.9 maintain action potential propagation in response to prolonged stimulation [53,54]. On the other hand, Ca2+ entry through VGCCs initiates several intracellular signaling cascades (such as cAMP, PKC, and MAPKs) that regulate ion channel function, trafficking, and gene expression [55].
Action potentials conducted along dorsal root axons primarily terminate in laminae I and II of the dorsal horn, forming synapses with second-order projection neurons and multiple interneuron types (Figure 2) [56]. Neurotransmission at these sites involves the release of excitatory neurotransmitters and neuromodulators (including glutamate, substance P, and CGRP) that modulate postsynaptic excitability and enhance nociceptive signal transmission to ascending spinal pathways such as the spinothalamic, spinoreticular, and spinoparabrachial tracts [57]. These ascending fibers project pain-related signals to a range of supraspinal structures (such as the thalamus, parabrachial nucleus, periaqueductal gray, amygdala, and somatosensory cortices), facilitating processing of the sensory-discriminative and affective-emotional components of pain [58].
A salient feature of nociceptors is their capacity for plasticity, a property that enables them to modulate their excitability and response thresholds in the context of inflammation, injury, or disease [59]. This neuroplastic adaptation constitutes the basis of peripheral sensitization, a process whereby exposure to inflammatory mediators such as PGE2, bradykinin, TNF-α, IL-1β, and NGF enhances the excitability of nociceptors, thereby promoting the emergence of hyperalgesia and allodynia in the context of injury or inflammation [60,61,62,63,64]. Sensitization involves post-translational modifications that enhance or modulate ion channel function (contributing to increased neuronal excitability) via phosphorylation by intracellular signaling cascades including PKA, PKC, and MAPKs [64] and longer-term transcriptional reprogramming that results in the upregulation of several pro-nociceptive genes [65]. In particular, NGF-TrkA signaling exerts potent effects on nociceptor gene expression, trafficking of ion channels to the membrane, and cytoskeletal dynamics, thereby contributing to both the acute sensitization and chronic hyperexcitability of these neurons [66].
Finally, under pathological conditions (particularly in neuropathic pain), nociceptors can undergo marked phenotypic transformations, leading to the generation of ectopic and spontaneous activity [67]. Additionally, interactions between injured nociceptors and immune elements, such as macrophages, T cells, and satellite glial cells, result in the establishment of a pro-inflammatory microenvironment that evokes nociceptive signaling [68]. This neuroimmune crosstalk is increasingly recognized as a key driver of pain chronification and involves modulation of cytokine release, chemokine signaling, and ion channel expression [69].
In summary, nociceptors comprise a highly specialized subset of peripheral sensory neurons, functionally specialized to detect, transduce, and transmit a wide array of potentially damaging stimuli [32]. Their molecular and functional plasticity not only facilitates adaptive responses within the pain pathway but also plays a central role in the development and maintenance of chronic pain states [59]. Advances in neurobiology, molecular genetics, and translational research are progressively clarifying the biology of nociceptors, thereby facilitating the design of highly effective analgesic strategies [70,71]. Defining nociceptor-specific signaling pathways and understanding their modulation in pathological contexts remain pivotal challenges in the effort to mitigate chronic pain and improve patient outcomes [72].

3. Structural and Molecular Changes of Nociceptors Following an Injury

Tissue injury induces a series of structural and molecular alterations in nociceptors. These alterations constitute an adaptive response intended to preserve tissue integrity and promote healing [73]; however, when dysregulated, they may facilitate the transition from acute to chronic pain [74]. These changes are not passive ramifications of injury, but constitute dynamic responses orchestrated by intrinsic cellular programs and extrinsic signals from the surrounding microenvironment [40].

3.1. Molecular-Level Transformations

In the peripheral axon, injured nociceptors initiate a regenerative response characterized by cytoskeletal remodeling, neurite outgrowth, and remodeling of target-specific axonal projections. From a molecular perspective, one of the earliest and most robust hallmarks of this process is the upregulation of a distinct set of regeneration-associated genes (RAGs) [75], like activating transcription factor 3 (ATF3) [76], small proline-rich protein 1A (Sprr1a) [77], growth-associated protein 43 (GAP-43) [78], and the transcription factor c-Jun [79]. These RAGs coordinate a transcriptional program that promotes axonal sprouting and structural reorganization essential for the extension of new growth cones and the establishment of novel axonal branches [75]. GAP-43 plays an essential role in modulating the dynamics of the cytoskeleton by interacting with actin filaments and regulating membrane plasticity at the growth cone, thereby facilitating axonal elongation [80]. In the same way, c-Jun acts as a master regulator of gene expression in injured neurons, coordinating pathways that govern cell survival, cytoskeletal remodeling, and growth cone formation [81,82,83]. Sprr1a, another key RAG, contributes to cytoskeletal stability and promotes neurite extension by interacting with intermediate filaments and actin networks [84].
There is a notable shift in the expression profile of integrins and cell adhesion molecules (CAMs), like neural cell adhesion molecule (NCAM) and L1 cell adhesion molecule (L1CAM) [85,86]. These adhesion molecules mediate some interactions between regenerating axons and the extracellular matrix (ECM) [87], as well as with surrounding Schwann cells and other non-neuronal elements in the peripheral nerve environment [88]. The altered expression and glycosylation patterns of NCAM and L1CAM strengthen axonal attachment and promote neurite outgrowth by activating intracellular signaling cascades involving focal adhesion kinase (FAK) and integrin-linked kinase (ILK) [89,90]. These kinases regulate cytoskeletal dynamics via downstream effectors such as the PI3K/Akt and MAPK/ERK pathways, which are essential for growth cone motility and directional guidance [89,90]. Moreover, the ECM itself undergoes remodeling after injury, with increased deposition of laminin, fibronectin, and tenascin-C, which serve as permissive substrates for axonal growth [91,92]. The interaction of integrins with ECM components not only provides mechanical support but also triggers intracellular signaling that strengthens regenerative capacity [93,94]. Schwann cells play a key role in this process by secreting neurotrophic factors, such as NGF and brain-derived neurotrophic factor (BDNF), which further stimulate RAG expression and cytoskeletal remodeling in nociceptors [95,96].
Recent advances in single-cell transcriptomics have substantially expanded our understanding of the cellular and molecular heterogeneity present within injured nociceptors. In particular, the application of single-cell RNA sequencing (scRNA-seq) has enabled the high-resolution characterization of transcriptional diversity across individual nociceptive neurons following injury [97]. These studies have shown the existence of distinct subpopulations of nociceptors, each characterized by unique expression profiles of RAGs, as well as varying capacities for axonal regeneration [98,99]. For instance, landmark investigations have demonstrated that injury-induced transcriptional responses are not uniform across all sensory neurons but instead are stratified according to subtype-specific molecular programs. This fact has allowed for the identification of discrete molecular signatures associated with axonal regeneration, inflammatory signaling pathways, and pain hypersensitivity [100,101].
Simultaneously, injury elicits pronounced changes in the expression and function of several ion channels in nociceptors, which critically contribute to the emergence of hyperexcitability and the maintenance of persistent pain states [102]. A significant feature of this plasticity is the upregulation and altered localization of VGSCs (Nav1.3, Nav1.7, Nav1.8, and Nav1.9). Nav1.3, typically absent in adult peripheral neurons, is re-expressed de novo following nerve injury and exhibits rapid repriming kinetics that facilitate high-frequency firing [103]. Nav1.7, a threshold channel crucial for amplifying subthreshold depolarizations, becomes overexpressed, thereby lowering the threshold for action potential initiation [104]. Nav1.8 and Nav1.9, mostly expressed in nociceptors, contribute to sustained depolarizing currents and are resistant to inactivation, promoting ectopic firing [105,106]. These sodium channelopathies collectively enhance excitability and underlie spontaneous discharges observed in neuropathic pain. Concomitant changes are shown in the expression of K+ channels (VGKCs), most notably a downregulation of delayed rectifier channels such as Kv1.2, which under normal conditions contribute to membrane repolarization and the termination of action potentials [107]. The reduction in outward K+ currents prolongs depolarization phases, thus facilitating aberrant firing patterns [108]. Ca2+ channel expression is also perturbed, particularly with an upregulation of VGCCs (like Cav2.2), which are integral to presynaptic Ca2+ influx and neurotransmitter vesicle exocytosis at central terminals in the dorsal horn [109]. The enhanced activity of Cav2.2 channels leads to increased release of excitatory neurotransmitters such as glutamate and substance P, amplifying nociceptive signaling in the CNS [110].
Optogenetic methodologies have become indispensable for dissecting the functional architecture of nociceptive circuits, owing to their exquisite spatial, temporal, and genetic precision [111,112]. In the optogenetic domain, targeted expression of excitatory opsins in genetically defined nociceptor subsets allows millisecond-scale control over action-potential firing. Illumination with the adequate wavelength can elicit or suppress pain-related behaviors in freely moving mice, establishing causal links between specific neuron populations and discrete components of the nociceptive experience [113]. Some studies utilized ChR2 in Nav1.7, Nav1.8, and Nav1.9-Cre-positive neurons to demonstrate that activation of peptidergic afferents is sufficient to trigger pain behaviors without concomitant tissue injury, thereby separating nociceptor activity from inflammatory confounds [114,115].
Additionally, TRP channels (especially TRPV1, TRPA1, and TRPM8) are critically involved in the modulation of sensory neuron responsiveness to thermal, chemical, and mechanical stimuli [116]. TRPV1, activated by thermal and chemical stimuli (such as capsaicin), undergoes sensitization via phosphorylation by kinases, including PKC and PKA, resulting in a lowered activation threshold [117]. TRPA1, activated by ROS and various environmental irritants, is likewise sensitized and frequently co-expressed with TRPV1, contributing to the development of mechanical and cold hyperalgesia [118]. TRPM8, a critical mediator of cold sensation, may also be upregulated under some pathological conditions, thereby contributing to the development of cold allodynia [45]. Chemogenetic strategies deploy engineered G-protein–coupled receptors (DREADDs) that respond only to otherwise inert ligands such as clozapine-N-oxide (CNO) or the more pharmacologically stable compound deschloroclozapine (DCZ) [119,120]. Some studies showed that chemogenetic silencing of TRPV1-lineage afferents attenuates inflammatory hyperalgesia, whereas their activation exacerbates mechanical allodynia, thereby highlighting the sufficiency and necessity of these neurons in driving persistent pain states [121].
These ion channel modifications are driven by a milieu of pro-inflammatory mediators released in the injured microenvironment. Pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, along with neurotrophic factors (such as NGF) and lipid mediators like prostaglandins and bradykinin, bind to their respective receptors on nociceptor terminals [122,123,124,125,126]. These interactions initiate a cascade of intracellular signaling events, mainly involving MAPKs, PI3K/Akt, and PKC pathways [122,123,124,125,126]. All of these signaling cascades regulate transcriptional programs through activation of nuclear transcription factors like NF-κB and CREB, ultimately promoting the expression and post-translational modification of ion channels and receptors [127,128]. Moreover, these pathways influence axonal transport and membrane trafficking of several channel proteins, further enhancing nociceptor sensitivity and contributing to the persistent nature of chronic pain states [129].
Mitochondrial dynamics are also profoundly influenced by injury. Evidence suggests a shift toward increased mitochondrial fission, a process that facilitates the redistribution and transport of mitochondria along the axon [130]. This adaptation ensures the localized availability of ATP and other metabolic substrates at sites of axonal outgrowth and repair [131]. Enhanced mitochondrial transport from the soma to the distal axon terminals supports not only regenerative efforts but also sustains heightened synaptic activity, which is a hallmark of chronic pain states [132]. Moreover, injury-induced perturbations in the endoplasmic reticulum have been implicated in the pathophysiology of chronic nociception. Sustained endoplasmic reticulum stress induces the activation of the unfolded protein response (UPR), a fundamentally conserved cellular process responsible for the restoration [133]. Chronic activation of the UPR can result in maladaptive consequences, such as the modulation of ion channel expression, the induction of pro-inflammatory signaling cascades, and ultimately, the perpetuation of pain hypersensitivity [134,135,136].
Finally, the long-term persistence of these structural and functional changes in nociceptors is reinforced by epigenetic modifications (Figure 3), which function as reversible mechanisms of gene regulation without altering the DNA sequence [137]. These modifications include histone acetylation, DNA methylation, and the modulatory influence of ncRNAs, all of which play key roles in driving the epigenetic reprogramming of nociceptor identity and function [137]. These epigenetic changes serve as a molecular interface through which several extracellular signals are transduced into stable changes in chromatin architecture and transcriptional output.
One of the earliest epigenetic responses involves histone acetylation. Activated transcription factors (such as CREB and NF-κB) recruit histone acetyltransferases (HATs) like p300/CBP to the promoters and enhancers of some pain-relevant genes, including Scn9A, Scn10A, Trpv1, Bdnf, and Fos [138]. Concomitantly, a reduction in the expression or activity of histone deacetylases (HDACs), including HDAC1, HDAC2, HDAC4, and HDAC5, has been observed in injured nociceptors [139]. In parallel with several histone modifications, DNA methylation plays a key role in the epigenetic reprogramming of injured nociceptors [140]. In rodent models of nerve injury, errant DNA methylation patterns have been observed, including hypermethylation of CpG islands in the promoter regions of genes encoding certain VGKCs (Kcna2, Kcnq2, and Kcnd2) [141]. This leads to reduced gene expression, impairing membrane repolarization capacity, and enhancing potential firing. Conversely, hypomethylation of pro-inflammatory or pro-nociceptive genes (such as Trpa1, Il-6, and Atf3) results in their overexpression [142]. This selective demethylation is often facilitated by TET family enzymes, mainly TET1 and TET3, which catalyze the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine, initiating active DNA demethylation [143].
Another key layer of epigenetic regulation in injured nociceptors involves ncRNAs, miRNAs, lncRNAs, and circRNAs. For instance, miR-124 is downregulated in nociceptors after nerve injury, leading to disinhibition of key components of inflammatory pathways like TRAF6 and STAT3, and promoting the transcription of many pain-related genes [144]. In contrast, miR-21 is upregulated and contributes to nociceptor sensitization by targeting Pten and Spry2, negative regulators of MAPK signaling [145]. lncRNAs add a further dimension of complexity. The Kcna2 antisense RNA, transcribed from the opposite strand of the Kcna2 gene, can interact directly with the corresponding mRNA or genomic DNA to repress transcription. This downregulation leads to a significant reduction in VGKCs activity, thereby enhancing neuronal excitability [146]. Moreover, lncRNAs like Neat1 and MALAT1 function as scaffolds for chromatin-modifying complexes. Neat1 facilitates the recruitment of EZH2 to specific gene promoters, where it mediates the trimethylation of H3K27me3, thereby contributing to transcriptional silencing [147]. These interactions silence anti-nociceptive or homeostatic genes, further cementing the chronic pain phenotype.

3.2. Structural-Level Transformations

The regenerative process involves extensive cytoskeletal remodeling, including the dynamic assembly and alignment of microtubules and actin filaments [148], which confer mechanical stability and spatial guidance for advancing growth cones [149]. Microtubule-associated proteins (MAPs), such as MAP1B and τ, undergo phosphorylation changes that reconfigure microtubule dynamics, facilitating axonal elongation and branching [150]. Actin cytoskeleton remodeling is regulated by several Rho GTPases (e.g., RhoA, Rac1, and Cdc42), which orchestrate growth cone dynamics and directional guidance by modulating cycles of actin polymerization and depolymerization [151].
The surrounding non-neuronal cellular milieu exerts a profound influence on the initiation, maintenance, and exacerbation of neuroplastic and nociceptive processes following peripheral nerve injury (Figure 4) [152]. Schwann cells, in the peripheral nerve, and satellite glial cells (SGCs) in the DRG, undergo distinct phenotypic and functional reprogramming in response to injury, a process collectively known as reactive gliosis [153,154]. These adaptations are characterized by cellular proliferation and hypertrophy, increased expression of glial fibrillary acidic protein (GFAP), and augmented production and release of various pro-inflammatory and neuroactive mediators [155,156,157]. One of the distinguishing characteristics of this reactive phenotype is the increased expression of connexin 43 (Cx43), a gap junction protein that orchestrates direct cytoplasmic exchange between adjacent glial cells [158]. The upregulation of Cx43 reinforces intercellular coupling, thus synchronizing glial activity and amplifying neuroinflammatory signaling across the glial network [159]. This process not only supports the spread of Ca2+ waves and ATP-mediated signaling but also facilitates the paracrine and autocrine dissemination of some pro-inflammatory cytokines (such as IL-1β, IL-6, and TNF-α) [160]. Concurrently, Schwann cells and SGCs increase their release of neurotrophic factors, including BDNF, NGF, and glial cell line-derived neurotrophic factor (GDNF). Each of these factors exerts different effects on nociceptor excitability and plasticity [161].
Within the DRG, injury-induced alterations extend beyond peripheral axonal damage and encompass significant modifications in the somatic compartment of nociceptive neurons [162]. One of the earliest and most prominent cellular responses observed in the soma is a transformation in nuclear morphology. Following injury, the neuronal nucleus usually exhibits enhanced euchromatinization (Figure 3), evidenced by a relaxed and decondensed chromatin architecture [163,164]. This reflects an upregulation of transcriptional activity, as euchromatin is associated with an open chromatin state that enables active gene expression. Such transcriptional activation likely underlies the increased production of mRNAs required for both reparative and maladaptive cellular programs, including those associated with neuroinflammation and pain sensitization [165,166]. Simultaneously with chromatin remodeling, nucleolar hypertrophy is usually observed following peripheral nerve injury. The nucleolus, being the main site of rRNA transcription and ribosome assembly, shows morphological enlargement and upregulated function [167]. This morphological change reflects a substantial increase in ribosomal biogenesis, which is consistent with the heightened demand for protein synthesis necessary for axonal regeneration, cytoskeletal remodeling, and the synthesis of signaling molecules involved in nociceptive plasticity [168].
Centrally, nociceptive terminals located within the dorsal horn of the spinal cord undergo profound structural, molecular, and synaptic remodeling in response to peripheral nerve injury. One of the hallmark features of this reorganization is the aberrant sprouting of nociceptive C-fiber afferents into the dorsal horn laminae that are typically innervated by low-threshold mechanoreceptors [169]. Following injury, C-fiber terminals may extend into deeper laminae, such as lamina III, an area generally associated with innocuous tactile input conveyed by myelinated Aβ fibers [170]. This ectopic innervation enables nociceptors to form de novo synaptic contacts with interneurons and projection neurons that are not typically involved in pain processing under physiological conditions [171]. The resulting convergence of nociceptive and mechanoreceptive afferents onto common postsynaptic targets constitutes a critical mechanism underlying mechanical allodynia [172]. Injured nociceptors contribute significantly to this hyperexcitable state by releasing a suite of pro-nociceptive neurotransmitters and neuromodulators, such as glutamate, substance P, and CGRP [173]. These molecules promote central sensitization, a pathophysiological process characterized by increased excitability and synaptic efficacy of dorsal horn neurons [174].
Taken together, the structural transformations of nociceptors following injury are extensive and involve remodeling at the molecular and cellular levels. These changes extend from the peripheral terminals through the axon and soma to the central terminals in the spinal cord, forming a continuum of plasticity that supports the transition from acute to chronic pain. Understanding the precise mechanisms that govern these structural alterations holds promise for the development of targeted therapeutic strategies aimed at reversing or preventing pathological pain states.

4. Conclusions

In summary, peripheral nerve injury triggers many structural and molecular modifications in nociceptors that profoundly alter their functional properties and contribute to the pathogenesis of chronic pain. At the structural level, injury induces axonal sprouting, synaptic reorganization, and phenotypic shifts in nociceptors, including altered soma size and changes in connectivity with spinal interneurons. These changes are accompanied by a dynamic molecular landscape characterized by transcriptional reprogramming, epigenetic remodeling, and post-transcriptional regulation.
The molecular and cellular changes initiated by peripheral nerve damage extend beyond the acute phase of nociceptor sensitization, playing integral roles in the maintenance and amplification of chronic pain states. These injury-induced signatures, which encompass diverse classes of transcripts, proteins, post-translational modifications, and epigenetic marks, represent a rich reservoir of candidate biomarkers and actionable therapeutic targets with substantial translational potential [31]. Rather than viewing these molecular profiles solely as correlates of disease, emerging evidence suggests they might function as mechanistic drivers of pain chronification and as predictive indicators of therapeutic responsiveness.
Transcriptional reprogramming in injured nociceptors can be quantitatively profiled using many RNA sequencing approaches [100,101]. Such gene expression signatures have already begun to distinguish neuropathic pain subtypes linked to specific etiologies, such as diabetic neuropathy, traumatic nerve injury, or chemotherapy-induced peripheral neuropathy [175]. Epigenetic landscapes offer an additional molecular framework that could serve as reliable biomarkers of chronic pain states [176].
In addition to their diagnostic utility, these molecular profiles offer therapeutic leverage points. One promising avenue involves the modulation of ion channel expression and function. Given the key role of VGSCs, VGCCs, and TRP channels in regulating nociceptor excitability, pharmacological agents or gene-silencing approaches (such as siRNAs, antisense oligonucleotides, or CRISPR-Cas-based repressors) targeting these channels are under intense investigation [177]. Crucially, the specificity of these therapeutic interventions might be enhanced through integration with many molecular diagnostic approaches that confirm the upregulation of the corresponding targets in a given patient.
Similarly, the disruption of several pro-inflammatory signaling pathways constitutes an additional therapeutic strategy grounded in molecular pathology. Small molecule inhibitors of cytokine receptors (such as IL-1βR and TNFR1), chemokine antagonists (such as CCR2 and CXCR3), or intracellular kinases (e.g., MAPK, JNK, and IKKβ) have shown efficacy in preclinical models and are advancing through various stages of clinical development [178,179,180]. These interventions are designed to disrupt the pathological neuroimmune crosstalk that sustains nociceptor hyperexcitability and glial activation within the injured nervous system.
Epigenetic therapeutics constitute a key innovative frontier. HDAC inhibitors, DNA methyltransferase inhibitors, and RNA-based epigenetic modulators have demonstrated the capacity to reverse injury-induced transcriptional changes in nociceptors and restore homeostatic gene expression patterns. Such pharmacological agents, though still largely experimental in the context of pain, might offer the potential to “reset” nociceptor phenotype toward a non-sensitized state [181,182]. On the other hand, the development of targeted delivery systems (e.g., nanoparticle-based vectors or viral gene delivery platforms) might enable tissue- or cell-specific epigenetic interventions, thereby minimizing systemic side effects [183,184,185].
Importantly, these molecular strategies must be integrated into a framework of precision medicine. Chronic pain is a heterogeneous condition with many underlying mechanisms, and the identification of molecular endophenotypes within patient populations is essential for matching individuals with the most appropriate therapeutic modality [186]. Molecular biomarkers not only enable stratification of patients into mechanistically distinct subgroups but may also provide dynamic indicators of treatment efficacy, allowing real-time monitoring and adjustment of therapeutic regimens [187]. In this context, the convergence of molecular biology, systems neuroscience, and clinical pain research generates unprecedented opportunities. Multi-omics integration (combining transcriptomic, epigenomic, proteomic, and metabolomic data) can yield holistic disease signatures that more accurately reflect the complexity of chronic pain states. Coupled with machine learning and bioinformatics tools, these approaches may uncover latent molecular patterns that escape detection through conventional analyses.
Together, these findings illustrate that the nociceptor is not a passive conduit for pain signals but an active participant in the development of chronic pain through injury-induced neuroplasticity. Future research should focus on identifying the temporal sequence of these changes, their cell-type specificity, and their reversibility, which will be essential for the design of precision medicine approaches tailored to individual pain phenotypes.
Finally, this review offers a novel contribution by systematically delineating injury-induced structural and molecular adaptations that are specific to nociceptors. In contrast to previous studies that have focused on broader neuronal or systemic responses to injury, this article emphasizes the cellular remodeling and transcriptional reprogramming occurring within nociceptors. By providing a focused synthesis of these neuron-specific modifications, this review refines current understanding of pain mechanisms.

Funding

This research received no external funding.

Data Availability Statement

Not applicable. No new data were generated.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ASICAcid-sensing ion channel
ATF3Activating transcription factor 3
ATPAdenosine triphosphate
BDNFBrain-derived neurotrophic factor
CAMCell adhesion molecule
CaMKCalcium/calmodulin-dependent protein kinase
cAMPCyclic adenosine monophosphate
Cav2.2Voltage-gated calcium channel, subtype 2.2
CCL2Chemokine (C-C motif) ligand 2
CCR2C-C Motif chemokine receptor 2
Cdc42Cell division cycle 42
CGRPCalcitonin gene-related peptide
ChR2Channelrhodopsin-2
circRNACircular RNA
c-JunCellular Jun (a component of the AP-1 transcription factor complex)
CNOClozapine-N-oxide
CNSCentral nervous system
CREBcAMP response element-binding protein
CRISPRClustered regularly interspaced short palindromic repeats
CXCR2C-X-C Motif chemokine receptor 2
Cx43Connexin 43
DCZDeschloroclozapine
DNADeoxyribonucleic acid
DREADDDesigner receptor exclusively activated by designer drugs
DRGDorsal root ganglion
ECMExtracellular matrix
ERKExtracellular signal-regulated kinase
EZH2Enhancer of zeste homolog 2
FAKFocal adhesion kinase
GAP-43Growth-associated protein 43
GCN5General control non-derepressible 5
GDNFGlial cell line-derived neurotrophic factor
GFAPGlial Fibrillary Acidic Protein
H3K27me3Trimethylation of lysine 27 on histone H3
HATHistone acetyltransferase
HDACHistone deacetylase
HDAC1Histone deacetylase 1
HDAC2Histone deacetylase 2
HDAC4Histone deacetylase 4
HDAC5Histone deacetylase 5
IASPInternational Association for the Study of Pain
IKKβIκB kinase beta
IL-1βInterleukin 1 beta
IL-1βRInterleukin 1 beta receptor
IL-6Interleukin 6
ILKIntegrin-linked kinase
JNKc-Jun N-terminal kinase
Kcna2Potassium voltage-gated channel subfamily A member 2
Kcnd2Potassium voltage-gated channel subfamily D member 2
Kcnq2Potassium voltage-gated channel subfamily Q member 2
L1CAML1 cell adhesion molecule
lncRNALong non-coding RNA
MALAT1Metastasis-associated lung adenocarcinoma transcript 1
MAPMicrotubule-associated protein
MAP1BMicrotubule-associated protein 1B
MAPKMitogen-activated protein kinase
miRNAMicroRNA
mRNAMessenger ribonucleic acid
Nav1.7Voltage-gated sodium channel, subtype 1.7
Nav1.8Voltage-gated sodium channel, subtype 1.8
Nav1.9Voltage-gated sodium channel, subtype 1.9
NCAMNeural cell adhesion molecule
ncRNANon-coding RNA
Neat1Nuclear paraspeckle assembly transcript 1
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NGFNerve growth factor
P2XPurinergic receptor P2X
P2X3Purinergic Receptor P2X, ligand-gated ion channel 3
p300/CBPE1A binding protein p300/CREB-binding protein
PGE2Prostaglandin E2
PI3K/AktPhosphoinositide 3-kinase/protein kinase B
PiezoPiezo-type mechanosensitive ion channel
Piezo1Piezo-type mechanosensitive ion channel 1
Piezo2Piezo-type mechanosensitive ion channel 2
PKAProtein kinase A
PKCProtein kinase C
PLCPhospholipase C
PtenPhosphatase and tensin homolog
Rac1Ras-related C3 botulinum toxin substrate 1
RAGRegeneration-associated gene
Rho GTPaseRas homolog family of GTPases
RhoARas homolog gene family member A
ROSReactive oxygen species
rRNARibosomal ribonucleic acid
scRNA-seqSingle-cell RNA-sequencing
SGCSatellite glial cell
Sprr1aSmall proline-rich protein 1A
Spry2Sprouty RTK signaling antagonist 2
STAT3Signal transducer and activator of transcription 3
TETTen-eleven translocation (dioxygenase family)
TET1Ten-eleven translocation methylcytosine dioxygenase 1
TET3Ten-eleven translocation methylcytosine dioxygenase 3
TGTrigeminal ganglion
TNF-αTumor necrosis factor alpha
TNFR1Tumor necrosis factor receptor 1
TRAF6TNF receptor-associated factor 6
TrkATropomyosin receptor kinase A
TRPTransient receptor potential
TRPA1Transient receptor potential ankyrin 1
TRPM8Transient receptor potential melastatin 8
TRPV1Transient receptor potential vanilloid 1
UPRUnfolded protein response
VGCCVoltage-gated calcium channel
VGKCVoltage-gated potassium channel
VGSCVoltage-gated sodium channel

References

  1. Raja, S.N.; Carr, D.B.; Cohen, M.; Finnerup, N.B.; Flor, H.; Gibson, S.; Keefe, F.J.; Mogil, J.S.; Ringkamp, M.; Sluka, K.A.; et al. The revised International Association for the Study of Pain definition of pain: Concepts, challenges, and compromises. Pain 2020, 161, 1976–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Vader, K.; Bostick, G.P.; Carlesso, L.C.; Hunter, J.; Mesaroli, G.; Perreault, K.; Tousignant-Laflamme, Y.; Tupper, S.; Walton, D.M.; Wideman, T.H.; et al. The Revised IASP Definition of Pain and Accompanying Notes: Considerations for the Physiotherapy Profession. Physiother. Can. 2021, 73, 103–106. [Google Scholar] [CrossRef]
  3. Malfliet, A.; Coppieters, I.; Van Wilgen, P.; Kregel, J.; De Pauw, R.; Dolphens, M.; Ickmans, K. Brain changes associated with cognitive and emotional factors in chronic pain: A systematic review. Eur. J. Pain 2017, 21, 769–786. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, G.I.; Neumeister, M.W. Pain: Pathways and physiology. Clin. Plast. Surg. 2020, 47, 173–180. [Google Scholar] [CrossRef]
  5. Li, W.; Liu, P.; Hu, Y.; Meng, J. Pain Modulates Responses to Emotional Stimuli. Front. Psychol. 2020, 11, 595987. [Google Scholar] [CrossRef]
  6. Nikolenko, V.N.; Shelomentseva, E.M.; Tsvetkova, M.M.; Abdeeva, E.I.; Giller, D.B.; Babayeva, J.V.; Achkasov, E.E.; Gavryushova, L.V.; Sinelnikov, M.Y. Nociceptors: Their Role in Body’s Defenses, Tissue Specific Variations and Anatomical Update. J. Pain Res. 2022, 15, 867–877. [Google Scholar] [CrossRef]
  7. Middleton, S.J.; Barry, A.M.; Comini, M.; Li, Y.; Ray, P.R.; Shiers, S.; Themistocleous, A.C.; Uhelski, M.L.; Yang, X.; Dougherty, P.M.; et al. Studying human nociceptors: From fundamentals to clinic. Brain 2021, 144, 1312–1335. [Google Scholar] [CrossRef] [PubMed]
  8. McEntire, D.M.; Kirkpatrick, D.R.; Dueck, N.P.; Kerfeld, M.J.; Smith, T.A.; Nelson, T.J.; Reisbig, M.D.; Agrawal, D.K. Pain transduction: A pharmacologic perspective. Expert Rev. Clin. Pharmacol. 2016, 9, 1069–1080. [Google Scholar] [CrossRef]
  9. Gascon, E.; Moqrich, A. Heterogeneity in primary nociceptive neurons: From molecules to pathology. Arch. Pharm. Res. 2010, 33, 1489–1507. [Google Scholar] [CrossRef]
  10. Hanč, P.; von Andrian, U.H. No pain, no gain—How nociceptors orchestrate tissue repair. Cell Res. 2024, 34, 673–674. [Google Scholar] [CrossRef]
  11. Price, T.J.; Dussor, G. Evolution: The advantage of ‘maladaptive’ pain plasticity. Curr. Biol. 2014, 24, R384–R386. [Google Scholar] [CrossRef]
  12. Alvarez, P.; Bogen, O.; Green, P.G.; Levine, J.D. Nociceptor Overexpression of NaV1.7 Contributes to Chronic Muscle Pain Induced by Early-Life Stress. J. Pain 2021, 22, 806–816. [Google Scholar] [CrossRef] [PubMed]
  13. Xiao, Y.; Barbosa, C.; Pei, Z.; Xie, W.; Strong, J.A.; Zhang, J.M.; Cummins, T.R. Increased Resurgent Sodium Currents in Nav1.8 Contribute to Nociceptive Sensory Neuron Hyperexcitability Associated with Peripheral Neuropathies. J. Neurosci. 2019, 39, 1539–1550. [Google Scholar] [CrossRef] [PubMed]
  14. Hoffmann, T.; Kistner, K.; Carr, R.W.; Nassar, M.A.; Reeh, P.W.; Weidner, C. Reduced excitability and impaired nociception in peripheral unmyelinated fibers from Nav1.9-null mice. Pain 2017, 158, 58–67. [Google Scholar] [CrossRef]
  15. Duan, J.H.; Hodgdon, K.E.; Hingtgen, C.M.; Nicol, G.D. N-type calcium current, Cav2.2, is enhanced in small-diameter sensory neurons isolated from Nf1+/- mice. Neuroscience 2014, 270, 192–202. [Google Scholar] [CrossRef]
  16. Voolstra, O.; Huber, A. Post-Translational Modifications of TRP Channels. Cells 2014, 3, 258–287. [Google Scholar] [CrossRef] [PubMed]
  17. Viana, F. Nociceptors: Thermal allodynia and thermal pain. Handb. Clin. Neurol. 2018, 156, 103–119. [Google Scholar]
  18. Li, R.; Ye, J.J.; Gan, L.; Zhang, M.; Sun, D.; Li, Y.; Wang, T.; Chang, P. Traumatic inflammatory response: Pathophysiological role and clinical value of cytokines. Eur. J. Trauma Emerg. Surg. 2024, 50, 1313–1330. [Google Scholar] [CrossRef]
  19. Uçeyler, N.; Schäfers, M.; Sommer, C. Mode of action of cytokines on nociceptive neurons. Exp. Brain Res. 2009, 196, 67–78. [Google Scholar] [CrossRef]
  20. Illias, A.M.; Gist, A.C.; Zhang, H.; Kosturakis, A.K.; Dougherty, P.M. Chemokine CCL2 and its receptor CCR2 in the dorsal root ganglion contribute to oxaliplatin-induced mechanical hypersensitivity. Pain 2018, 159, 1308–1316. [Google Scholar] [CrossRef]
  21. Wang, F.; Stefano, G.B.; Kream, R.M. Epigenetic modification of DRG neuronal gene expression subsequent to nerve injury: Etiological contribution to complex regional pain syndromes (Part I). Med. Sci. Monit. 2014, 20, 1067–1077. [Google Scholar] [PubMed]
  22. Wang, F.; Stefano, G.B.; Kream, R.M. Epigenetic modification of DRG neuronal gene expression subsequent to nerve injury: Etiological contribution to complex regional pain syndromes (Part II). Med. Sci. Monit. 2014, 20, 1188–1200. [Google Scholar]
  23. Testa, L.; Dotta, S.; Vercelli, A.; Marvaldi, L. Communicating pain: Emerging axonal signaling in peripheral neuropathic pain. Front. Neuroanat. 2024, 18, 1398400. [Google Scholar] [CrossRef] [PubMed]
  24. Smith, P.R.; Campbell, Z.T. RNA-binding proteins in pain. Wiley Interdiscip. Rev. RNA 2024, 15, e1843. [Google Scholar] [CrossRef]
  25. Pinho-Ribeiro, F.A.; Verri, W.A., Jr.; Chiu, I.M. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017, 38, 5–19. [Google Scholar] [CrossRef] [PubMed]
  26. Greco, R.; Tassorelli, C.; Sandrini, G.; Di Bella, P.; Buscone, S.; Nappi, G. Role of calcitonin gene-related peptide and substance P in different models of pain. Cephalalgia 2008, 28, 114–126. [Google Scholar]
  27. Techameena, P.; Feng, X.; Zhang, K.; Hadjab, S. The single-cell transcriptomic atlas iPain identifies senescence of nociceptors as a therapeutical target for chronic pain treatment. Nat. Commun. 2024, 15, 8585. [Google Scholar] [CrossRef]
  28. Miller, R.E.; Kim, Y.S.; Tran, P.B.; Ishihara, S.; Dong, X.; Miller, R.J.; Malfait, A.M. Visualization of Peripheral Neuron Sensitization in a Surgical Mouse Model of Osteoarthritis by In Vivo Calcium Imaging. Arthritis Rheumatol. 2018, 70, 88–97. [Google Scholar] [CrossRef]
  29. Kumar, P.A.; Stallman, J.; Kharbat, Y.; Hoppe, J.; Leonards, A.; Kerim, E.; Nguyen, B.; Adkins, R.L.; Baltazar, A.; Milligan, S.; et al. Chemogenetic Attenuation of Acute Nociceptive Signaling Enhances Functional Outcomes Following Spinal Cord Injury. J. Neurotrauma 2024, 41, 1060–1076. [Google Scholar] [CrossRef]
  30. Wang, Q.; Chen, Y.; Ding, H.; Cai, Y.; Yuan, X.; Lv, J.; Huang, J.; Huang, J.; Zhang, C.; Hong, Z.; et al. Optogenetic activation of mechanical nociceptions to enhance implant osseointegration. Nat. Commun. 2025, 16, 3093. [Google Scholar] [CrossRef]
  31. Berta, T.; Perrin, F.E.; Pertin, M.; Tonello, R.; Liu, Y.C.; Chamessian, A.; Kato, A.C.; Ji, R.R.; Decosterd, I. Gene Expression Profiling of Cutaneous Injured and Non-Injured Nociceptors in SNI Animal Model of Neuropathic Pain. Sci. Rep. 2017, 7, 9367. [Google Scholar] [CrossRef] [PubMed]
  32. Woolf, C.J.; Ma, Q. Nociceptors--noxious stimulus detectors. Neuron 2007, 55, 353–364. [Google Scholar] [CrossRef] [PubMed]
  33. Lacinova, L. Electrophysiology of nociception: Understanding of signaling pathways forms a basis for potential treatment. Pflugers Arch. 2022, 474, 365–366. [Google Scholar] [CrossRef] [PubMed]
  34. Motzkin, J.C.; Basbaum, A.I.; Crowther, A.J. Neuroanatomy of the nociceptive system: From nociceptors to brain networks. Int. Rev. Neurobiol. 2024, 179, 1–39. [Google Scholar]
  35. Todd, A.J. Neuronal circuitry for pain processing in the dorsal horn. Nat. Rev. Neurosci. 2010, 11, 823–836. [Google Scholar] [CrossRef]
  36. Raz, N.; Granovsky, Y.; Defrin, R. Investigating the neural processing of spatial summation of pain: The role of A-delta nociceptors. Exp. Brain Res. 2015, 233, 405–413. [Google Scholar] [CrossRef]
  37. Serra, J.; Collado, A.; Solà, R.; Antonelli, F.; Torres, X.; Salgueiro, M.; Quiles, C.; Bostock, H. Hyperexcitable C nociceptors in fibromyalgia. Ann. Neurol. 2014, 75, 196–208. [Google Scholar] [CrossRef]
  38. Emery, E.C.; Wood, J.N. Somatosensation a la mode: Plasticity and polymodality in sensory neurons. Curr. Opin. Physiol. 2019, 11, 29–34. [Google Scholar] [CrossRef]
  39. Kupari, J.; Ernfors, P. Molecular taxonomy of nociceptors and pruriceptors. Pain 2023, 164, 1245–1257. [Google Scholar] [CrossRef]
  40. Tracey, W.D., Jr. Nociception. Curr. Biol. 2017, 27, R129–R133. [Google Scholar] [CrossRef]
  41. Giniatullin, R. Ion Channels of Nociception. Int. J. Mol. Sci. 2020, 21, 3553. [Google Scholar] [CrossRef]
  42. Huang, J.; Korsunsky, A.; Yazdani, M.; Chen, J. Targeting TRP channels: Recent advances in structure, ligand binding, and molecular mechanisms. Front. Mol. Neurosci. 2024, 16, 1334370. [Google Scholar] [CrossRef]
  43. Benítez-Angeles, M.; Morales-Lázaro, S.L.; Juárez-González, E.; Rosenbaum, T. TRPV1: Structure, Endogenous Agonists, and Mechanisms. Int. J. Mol. Sci. 2020, 21, 3421. [Google Scholar] [CrossRef]
  44. Bautista, D.M.; Pellegrino, M.; Tsunozaki, M. TRPA1: A gatekeeper for inflammation. Annu. Rev. Physiol. 2013, 75, 181–200. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Y.; Mikrani, R.; He, Y.; Faran Ashraf Baig, M.M.; Abbas, M.; Naveed, M.; Tang, M.; Zhang, Q.; Li, C.; Zhou, X. TRPM8 channels: A review of distribution and clinical role. Eur. J. Pharmacol. 2020, 882, 173312. [Google Scholar] [CrossRef] [PubMed]
  46. Delmas, P.; Parpaite, T.; Coste, B. PIEZO channels and newcomers in the mammalian mechanosensitive ion channel family. Neuron 2022, 110, 2713–2727. [Google Scholar] [CrossRef] [PubMed]
  47. Szczot, M.; Nickolls, A.R.; Lam, R.M.; Chesler, A.T. The Form and Function of PIEZO2. Annu. Rev. Biochem. 2021, 90, 507–534. [Google Scholar] [CrossRef]
  48. Liu, H.; Hu, J.; Zheng, Q.; Feng, X.; Zhan, F.; Wang, X.; Xu, G.; Hua, F. Piezo1 Channels as Force Sensors in Mechanical Force-Related Chronic Inflammation. Front. Immunol. 2022, 13, 816149. [Google Scholar] [CrossRef]
  49. Storozhuk, M.; Cherninskyi, A.; Maximyuk, O.; Isaev, D.; Krishtal, O. Acid-Sensing Ion Channels: Focus on Physiological and Some Pathological Roles in the Brain. Curr. Neuropharmacol. 2021, 19, 1570–1589. [Google Scholar]
  50. Burnstock, G. Purinergic Mechanisms and Pain. Adv. Pharmacol. 2016, 75, 91–137. [Google Scholar]
  51. Docherty, R.J.; Farmer, C.E. The pharmacology of voltage-gated sodium channels in sensory neurones. Handb. Exp. Pharmacol. 2009, 194, 519–561. [Google Scholar]
  52. Dib-Hajj, S.D.; Yang, Y.; Black, J.A.; Waxman, S.G. The Na(V)1.7 sodium channel: From molecule to man. Nat. Rev. Neurosci. 2013, 14, 49–62. [Google Scholar] [CrossRef]
  53. Heinle, J.W.; Dalessio, S.; Janicki, P.; Ouyang, A.; Vrana, K.E.; Ruiz-Velasco, V.; Coates, M.D. Insights into the voltage-gated sodium channel, NaV1.8, and its role in visceral pain perception. Front. Pharmacol. 2024, 15, 1398409. [Google Scholar] [CrossRef]
  54. Zhang, X.; Hartung, J.E.; Gold, M.S. Persistent (Na v 1.9) sodium currents in human dorsal root ganglion neurons. Pain 2025, 166, 448–459. [Google Scholar] [CrossRef]
  55. Correa, B.H.M.; Moreira, C.R.; Hildebrand, M.E.; Vieira, L.B. The Role of Voltage-Gated Calcium Channels in Basal Ganglia Neurodegenerative Disorders. Curr. Neuropharmacol. 2023, 21, 183–201. [Google Scholar] [CrossRef]
  56. Tan, S.; Faull, R.L.M.; Curtis, M.A. The tracts, cytoarchitecture, and neurochemistry of the spinal cord. Anat. Rec. (Hoboken) 2023, 306, 777–819. [Google Scholar] [CrossRef]
  57. Willis, W.D. Nociceptive pathways: Anatomy and physiology of nociceptive ascending pathways. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1985, 308, 253–270. [Google Scholar]
  58. Yarmolinsky, D.A.; Zeng, X.; MacKinnon-Booth, N.; Greene, C.A.; Kim, C.; Cheng, Y.T.; Lenfers Turnes, B.; Woolf, C.J. Differential modification of ascending spinal outputs in acute and chronic pain states. Neuron 2025, 113, 1223–1239.e5. [Google Scholar] [CrossRef]
  59. Pace, M.C.; Passavanti, M.B.; De Nardis, L.; Bosco, F.; Sansone, P.; Pota, V.; Barbarisi, M.; Palagiano, A.; Iannotti, F.A.; Panza, E.; et al. Nociceptor plasticity: A closer look. J. Cell Physiol. 2018, 233, 2824–2838. [Google Scholar] [CrossRef]
  60. Ellis, A.; Bennett, D.L. Neuroinflammation and the generation of neuropathic pain. Br. J. Anaesth. 2013, 111, 26–37. [Google Scholar] [CrossRef]
  61. Hanč, P.; Messou, M.A.; Ajit, J.; von Andrian, U.H. Setting the tone: Nociceptors as conductors of immune responses. Trends Immunol. 2024, 45, 783–798. [Google Scholar] [CrossRef]
  62. Ronchetti, S.; Migliorati, G.; Delfino, D.V. Association of inflammatory mediators with pain perception. Biomed. Pharmacother. 2017, 96, 1445–1452. [Google Scholar] [CrossRef]
  63. Gold, M.S.; Gebhart, G.F. Nociceptor sensitization in pain pathogenesis. Nat. Med. 2010, 16, 1248–1257. [Google Scholar] [CrossRef]
  64. García-Domínguez, M. The Role of TNF-α in Neuropathic Pain: An Immunotherapeutic Perspective. Life 2025, 15, 785. [Google Scholar] [CrossRef]
  65. Cheng, J.K.; Ji, R.R. Intracellular signaling in primary sensory neurons and persistent pain. Neurochem. Res. 2008, 33, 1970–1978. [Google Scholar] [CrossRef]
  66. Mizumura, K.; Murase, S. Role of nerve growth factor in pain. Handb. Exp. Pharmacol. 2015, 227, 57–77. [Google Scholar]
  67. von Hehn, C.A.; Baron, R.; Woolf, C.J. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012, 73, 638–652. [Google Scholar] [CrossRef]
  68. Jain, A.; Gyori, B.M.; Hakim, S.; Jain, A.; Sun, L.; Petrova, V.; Bhuiyan, S.A.; Zhen, S.; Wang, Q.; Kawaguchi, R.; et al. Nociceptor-immune interactomes reveal insult-specific immune signatures of pain. Nat. Immunol. 2024, 25, 1296–1305. [Google Scholar] [CrossRef]
  69. Hore, Z.; Denk, F. Neuroimmune interactions in chronic pain—An interdisciplinary perspective. Brain Behav. Immun. 2019, 79, 56–62. [Google Scholar] [CrossRef]
  70. St John Smith, E. Advances in understanding nociception and neuropathic pain. J. Neurol. 2018, 265, 231–238. [Google Scholar] [CrossRef]
  71. Moran, M.M.; Szallasi, A. Targeting nociceptive transient receptor potential channels to treat chronic pain: Current state of the field. Br. J. Pharmacol. 2018, 175, 2185–2203. [Google Scholar] [CrossRef] [PubMed]
  72. Bublitz, V.; Ringat, T.; Jurth, C.; Lichtner, G.; von Dincklage, F. Performance of the Nociception Level Index and the PainSensor to predict and detect responsiveness to nociceptive procedures in critical care patients. J. Crit. Care 2025, 88, 155090. [Google Scholar] [CrossRef] [PubMed]
  73. Dubin, A.E.; Patapoutian, A. Nociceptors: The sensors of the pain pathway. J. Clin. Investig. 2010, 120, 3760–3772. [Google Scholar] [CrossRef]
  74. Walters, E.T. Nociceptors as chronic drivers of pain and hyperreflexia after spinal cord injury: An adaptive-maladaptive hyperfunctional state hypothesis. Front. Physiol. 2012, 3, 309. [Google Scholar] [CrossRef] [PubMed]
  75. Ma, T.C.; Willis, D.E. What makes a RAG regeneration associated? Front. Mol. Neurosci. 2015, 8, 43. [Google Scholar] [CrossRef]
  76. Cheng, Y.C.; Snavely, A.; Barrett, L.B.; Zhang, X.; Herman, C.; Frost, D.J.; Riva, P.; Tochitsky, I.; Kawaguchi, R.; Singh, B.; et al. Topoisomerase I inhibition and peripheral nerve injury induce DNA breaks and ATF3-associated axon regeneration in sensory neurons. Cell. Rep. 2021, 36, 109666. [Google Scholar] [CrossRef]
  77. Starkey, M.L.; Davies, M.; Yip, P.K.; Carter, L.M.; Wong, D.J.; McMahon, S.B.; Bradbury, E.J. Expression of the regeneration-associated protein SPRR1A in primary sensory neurons and spinal cord of the adult mouse following peripheral and central injury. J. Comp. Neurol. 2009, 513, 51–68. [Google Scholar] [CrossRef]
  78. Ondarza, A.B.; Ye, Z.; Hulsebosch, C.E. Direct evidence of primary afferent sprouting in distant segments following spinal cord injury in the rat: Colocalization of GAP-43 and CGRP. Exp. Neurol. 2003, 184, 373–380. [Google Scholar] [CrossRef]
  79. Son, S.J.; Lee, K.M.; Jeon, S.M.; Park, E.S.; Park, K.M.; Cho, H.J. Activation of transcription factor c-jun in dorsal root ganglia induces VIP and NPY upregulation and contributes to the pathogenesis of neuropathic pain. Exp. Neurol. 2007, 204, 467–472. [Google Scholar] [CrossRef]
  80. Chung, D.; Shum, A.; Caraveo, G. GAP-43 and BASP1 in Axon Regeneration: Implications for the Treatment of Neurodegenerative Diseases. Front. Cell Dev. Biol. 2020, 8, 567537. [Google Scholar] [CrossRef]
  81. Whitfield, J.; Neame, S.J.; Paquet, L.; Bernard, O.; Ham, J. Dominant-negative c-Jun promotes neuronal survival by reducing BIM expression and inhibiting mitochondrial cytochrome c release. Neuron 2001, 29, 629–643. [Google Scholar] [CrossRef] [PubMed]
  82. Oren, A.; Herschkovitz, A.; Ben-Dror, I.; Holdengreber, V.; Ben-Shaul, Y.; Seger, R.; Vardimon, L. The cytoskeletal network controls c-Jun expression and glucocorticoid receptor transcriptional activity in an antagonistic and cell-type-specific manner. Mol. Cell. Biol. 1999, 19, 1742–1750. [Google Scholar] [CrossRef]
  83. Ruff, C.A.; Staak, N.; Patodia, S.; Kaswich, M.; Rocha-Ferreira, E.; Da Costa, C.; Brecht, S.; Makwana, M.; Fontana, X.; Hristova, M.; et al. Neuronal c-Jun is required for successful axonal regeneration, but the effects of phosphorylation of its N-terminus are moderate. J. Neurochem. 2012, 121, 607–618. [Google Scholar] [CrossRef]
  84. Bonilla, I.E.; Tanabe, K.; Strittmatter, S.M. Small proline-rich repeat protein 1A is expressed by axotomized neurons and promotes axonal outgrowth. J. Neurosci. 2002, 22, 1303–1315. [Google Scholar] [CrossRef]
  85. Schäfer, M.K.; Frotscher, M. Role of L1CAM for axon sprouting and branching. Cell Tissue Res. 2012, 349, 39–48. [Google Scholar] [CrossRef] [PubMed]
  86. Quartu, M.; Serra, M.P.; Boi, M.; Ibba, V.; Melis, T.; Del Fiacco, M. Polysialylated-neural cell adhesion molecule (PSA-NCAM) in the human trigeminal ganglion and brainstem at prenatal and adult ages. BMC Neurosci. 2008, 9, 108. [Google Scholar] [CrossRef] [PubMed]
  87. Kozlova, I.; Sah, S.; Keable, R.; Leshchyns’ka, I.; Janitz, M.; Sytnyk, V. Cell Adhesion Molecules and Protein Synthesis Regulation in Neurons. Front. Mol. Neurosci. 2020, 13, 592126. [Google Scholar] [CrossRef]
  88. Spiegel, I.; Adamsky, K.; Eisenbach, M.; Eshed, Y.; Spiegel, A.; Mirsky, R.; Scherer, S.S.; Peles, E. Identification of novel cell-adhesion molecules in peripheral nerves using a signal-sequence trap. Neuron Glia Biol. 2006, 2, 27–38. [Google Scholar] [CrossRef]
  89. Weledji, E.P.; Assob, J.C. The ubiquitous neural cell adhesion molecule (N-CAM). Ann. Med. Surg. (Lond.) 2014, 3, 77–81. [Google Scholar] [CrossRef]
  90. Pusey, M.A.; Pace, K.; Fascelli, M.; Linser, P.J.; Steindler, D.A.; Galileo, D.S. Ectopic expression of L1CAM ectodomain alters differentiation and motility, but not proliferation, of human neural progenitor cells. Int. J. Dev. Neurosci. 2019, 78, 49–64. [Google Scholar] [CrossRef]
  91. Nelson, D.W.; Gilbert, R.J. Extracellular Matrix-Mimetic Hydrogels for Treating Neural Tissue Injury: A Focus on Fibrin, Hyaluronic Acid, and Elastin-Like Polypeptide Hydrogels. Adv. Healthc. Mater. 2021, 10, e2101329. [Google Scholar] [CrossRef]
  92. Yu, P.; Zhang, G.; Hou, B.; Song, E.; Wen, J.; Ba, Y.; Zhu, D.; Wang, G.; Qin, F. Effects of ECM proteins (laminin, fibronectin, and type IV collagen) on the biological behavior of Schwann cells and their roles in the process of remyelination after peripheral nerve injury. Front. Bioeng. Biotechnol. 2023, 11, 1133718. [Google Scholar] [CrossRef] [PubMed]
  93. Berti, C.; Nodari, A.; Wrabetz, L.; Feltri, M.L. Role of integrins in peripheral nerves and hereditary neuropathies. Neuromol. Med. 2006, 8, 191–204. [Google Scholar] [CrossRef] [PubMed]
  94. Myers, J.P.; Santiago-Medina, M.; Gomez, T.M. Regulation of axonal outgrowth and pathfinding by integrin-ECM interactions. Dev. Neurobiol. 2011, 71, 901–923. [Google Scholar] [CrossRef]
  95. Boyd, J.G.; Gordon, T. Neurotrophic factors and their receptors in axonal regeneration and functional recovery after peripheral nerve injury. Mol. Neurobiol. 2003, 27, 277–324. [Google Scholar] [CrossRef] [PubMed]
  96. Jessen, K.R.; Mirsky, R. The repair Schwann cell and its function in regenerating nerves. J. Physiol. 2016, 594, 3521–3531. [Google Scholar] [CrossRef]
  97. Jovic, D.; Liang, X.; Zeng, H.; Lin, L.; Xu, F.; Luo, Y. Single-cell RNA sequencing technologies and applications: A brief overview. Clin. Transl. Med. 2022, 12, e694. [Google Scholar] [CrossRef]
  98. Leone, C.M.; Lenoir, C.; van den Broeke, E.N. Assessing signs of central sensitization: A critical review of physiological measures in experimentally induced secondary hyperalgesia. Eur. J. Pain 2025, 29, e4733. [Google Scholar] [CrossRef]
  99. Hu, G.; Huang, K.; Hu, Y.; Du, G.; Xue, Z.; Zhu, X.; Fan, G. Single-cell RNA-seq reveals distinct injury responses in different types of DRG sensory neurons. Sci. Rep. 2016, 6, 31851. [Google Scholar] [CrossRef]
  100. Cooper, A.H.; Barry, A.M.; Chrysostomidou, P.; Lolignier, R.; Wang, J.; Redondo Canales, M.; Titterton, H.F.; Bennett, D.L.; Weir, G.A. Peripheral nerve injury results in a biased loss of sensory neuron subpopulations. Pain 2024, 165, 2863–2876. [Google Scholar] [CrossRef]
  101. Bolívar, S.; Sanz, E.; Ovelleiro, D.; Zochodne, D.W.; Udina, E. Neuron-specific RNA-sequencing reveals different responses in peripheral neurons after nerve injury. eLife 2024, 12, RP91316. [Google Scholar] [CrossRef] [PubMed]
  102. Waxman, S.G.; Zamponi, G.W. Regulating excitability of peripheral afferents: Emerging ion channel targets. Nat. Neurosci. 2014, 17, 153–163. [Google Scholar] [CrossRef] [PubMed]
  103. Waxman, S.G.; Kocsis, J.D.; Black, J.A. Type III sodium channel mRNA is expressed in embryonic but not adult spinal sensory neurons, and is reexpressed following axotomy. J. Neurophysiol. 1994, 72, 466–470. [Google Scholar] [CrossRef]
  104. Cai, W.; Zhao, Q.; Shao, J.; Zhang, J.; Li, L.; Ren, X.; Su, S.; Bai, Q.; Li, M.; Chen, X.; et al. MicroRNA-182 Alleviates Neuropathic Pain by Regulating Nav1.7 Following Spared Nerve Injury in Rats. Sci. Rep. 2018, 8, 16750. [Google Scholar] [CrossRef] [PubMed]
  105. Thakor, D.K.; Lin, A.; Matsuka, Y.; Meyer, E.M.; Ruangsri, S.; Nishimura, I.; Spigelman, I. Increased peripheral nerve excitability and local NaV1.8 mRNA up-regulation in painful neuropathy. Mol. Pain 2009, 5, 14. [Google Scholar] [CrossRef]
  106. Sidaway, P. Pain: Gain-of-function Nav1.9 mutations are associated with painful peripheral neuropathy. Nat. Rev. Neurol. 2014, 10, 306. [Google Scholar] [CrossRef]
  107. Fan, L.; Guan, X.; Wang, W.; Zhao, J.Y.; Zhang, H.; Tiwari, V.; Hoffman, P.N.; Li, M.; Tao, Y.X. Impaired neuropathic pain and preserved acute pain in rats overexpressing voltage-gated potassium channel subunit Kv1.2 in primary afferent neurons. Mol. Pain 2014, 10, 8. [Google Scholar] [CrossRef]
  108. Du, X.; Gamper, N. Potassium channels in peripheral pain pathways: Expression, function and therapeutic potential. Curr. Neuropharmacol. 2013, 11, 621–640. [Google Scholar] [CrossRef]
  109. Yang, J.; Xie, M.X.; Hu, L.; Wang, X.F.; Mai, J.Z.; Li, Y.Y.; Wu, N.; Zhang, C.; Li, J.; Pang, R.P.; et al. Upregulation of N-type calcium channels in the soma of uninjured dorsal root ganglion neurons contributes to neuropathic pain by increasing neuronal excitability following peripheral nerve injury. Brain Behav. Immun. 2018, 71, 52–65. [Google Scholar] [CrossRef]
  110. Antunes, F.T.T.; Campos, M.M.; Carvalho, V.P.R.; da Silva Junior, C.A.; Magno, L.A.V.; de Souza, A.H.; Gomez, M.V. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int. J. Mol. Sci. 2023, 24, 9223. [Google Scholar] [CrossRef]
  111. Emiliani, V.; Entcheva, E.; Hedrich, R.; Hegemann, P.; Konrad, K.R.; Lüscher, C.; Mahn, M.; Pan, Z.H.; Sims, R.R.; Vierock, J.; et al. Optogenetics for light control of biological systems. Nat. Rev. Methods Primers 2022, 2, 55. [Google Scholar] [CrossRef]
  112. Chen, W.; Li, C.; Liang, W.; Li, Y.; Zou, Z.; Xie, Y.; Liao, Y.; Yu, L.; Lin, Q.; Huang, M.; et al. The Roles of Optogenetics and Technology in Neurobiology: A Review. Front. Aging Neurosci. 2022, 14, 867863. [Google Scholar] [CrossRef] [PubMed]
  113. Mickle, A.D.; Gereau, R.W. 4th. A bright future? Optogenetics in the periphery for pain research and therapy. Pain 2018, 159 (Suppl. 1), S65–S73. [Google Scholar] [CrossRef]
  114. Daou, I.; Beaudry, H.; Ase, A.R.; Wieskopf, J.S.; Ribeiro-da-Silva, A.; Mogil, J.S.; Séguéla, P. Optogenetic Silencing of Nav1.8-Positive Afferents Alleviates Inflammatory and Neuropathic Pain. eNeuro 2016, 3, ENEURO.0140-15.2016. [Google Scholar] [CrossRef] [PubMed]
  115. Maruta, T.; Kouroki, S.; Kurogi, M.; Hidaka, K.; Koshida, T.; Miura, A.; Nakagawa, H.; Yanagita, T.; Takeya, R.; Tsuneyoshi, I. Comparison of Nocifensive Behavior in NaV1.7-, NaV1.8-, and NaV1.9-Channelrhodopsin-2 Mice by Selective Optogenetic Activation of Targeted Sodium Channel Subtype-Expressing Afferents. J. Neurosci. Res. 2024, 102, e25386. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, M.; Ma, Y.; Ye, X.; Zhang, N.; Pan, L.; Wang, B. TRP (transient receptor potential) ion channel family: Structures, biological functions and therapeutic interventions for diseases. Signal Transduct. Target Ther. 2023, 8, 261. [Google Scholar] [CrossRef]
  117. Shuba, Y.M. Beyond Neuronal Heat Sensing: Diversity of TRPV1 Heat-Capsaicin Receptor-Channel Functions. Front. Cell Neurosci. 2021, 14, 612480. [Google Scholar] [CrossRef]
  118. Meents, J.E.; Ciotu, C.I.; Fischer, M.J.M. TRPA1: A molecular view. J. Neurophysiol. 2019, 121, 427–443. [Google Scholar] [CrossRef]
  119. Campbell, E.J.; Marchant, N.J. The use of chemogenetics in behavioural neuroscience: Receptor variants, targeting approaches and caveats. Br. J. Pharmacol. 2018, 175, 994–1003. [Google Scholar] [CrossRef]
  120. Poth, K.M.; Texakalidis, P.; Boulis, N.M. Chemogenetics: Beyond Lesions and Electrodes. Neurosurgery 2021, 89, 185–195. [Google Scholar] [CrossRef]
  121. Hsiao, I.H.; Yen, C.M.; Hsu, H.C.; Liao, H.Y.; Lin, Y.W. Chemogenetics Modulation of Electroacupuncture Analgesia in Mice Spared Nerve Injury-Induced Neuropathic Pain through TRPV1 Signaling Pathway. Int. J. Mol. Sci. 2024, 25, 1771. [Google Scholar] [CrossRef] [PubMed]
  122. Cook, A.D.; Christensen, A.D.; Tewari, D.; McMahon, S.B.; Hamilton, J.A. Immune Cytokines and Their Receptors in Inflammatory Pain. Trends Immunol. 2018, 39, 240–255. [Google Scholar] [CrossRef]
  123. García-Domínguez, M. NGF in Neuropathic Pain: Understanding Its Role and Therapeutic Opportunities. Curr. Issues Mol. Biol. 2025, 47, 93. [Google Scholar] [CrossRef]
  124. Merighi, A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024, 14, 539. [Google Scholar] [CrossRef]
  125. Jang, Y.; Kim, M.; Hwang, S.W. Molecular mechanisms underlying the actions of arachidonic acid-derived prostaglandins on peripheral nociception. J. Neuroinflamm. 2020, 17, 30. [Google Scholar] [CrossRef] [PubMed]
  126. Mizumura, K.; Sugiura, T.; Katanosaka, K.; Banik, R.K.; Kozaki, Y. Excitation and sensitization of nociceptors by bradykinin: What do we know? Exp. Brain Res. 2009, 196, 53–65. [Google Scholar] [CrossRef]
  127. Kanngiesser, M.; Häussler, A.; Myrczek, T.; Küsener, N.; Lim, H.Y.; Geisslinger, G.; Niederberge, E.; Tegeder, I. Inhibitor kappa B kinase beta dependent cytokine upregulation in nociceptive neurons contributes to nociceptive hypersensitivity after sciatic nerve injury. J. Pain 2012, 13, 485–497. [Google Scholar] [CrossRef]
  128. Melemedjian, O.K.; Tillu, D.V.; Moy, J.K.; Asiedu, M.N.; Mandell, E.K.; Ghosh, S.; Dussor, G.; Price, T.J. Local translation and retrograde axonal transport of CREB regulates IL-6-induced nociceptive plasticity. Mol. Pain 2014, 10, 45. [Google Scholar] [CrossRef] [PubMed]
  129. Patodia, S.; Raivich, G. Role of transcription factors in peripheral nerve regeneration. Front. Mol. Neurosci. 2012, 5, 8. [Google Scholar] [CrossRef]
  130. Espinoza, N.; Papadopoulos, V. Role of Mitochondrial Dysfunction in Neuropathy. Int. J. Mol. Sci. 2025, 26, 3195. [Google Scholar] [CrossRef]
  131. Lim, T.K.; Rone, M.B.; Lee, S.; Antel, J.P.; Zhang, J. Mitochondrial and bioenergetic dysfunction in trauma-induced painful peripheral neuropathy. Mol. Pain 2015, 11, 58. [Google Scholar] [CrossRef] [PubMed]
  132. Silva Santos Ribeiro, P.; Willemen, H.L.D.M.; Eijkelkamp, N. Mitochondria and sensory processing in inflammatory and neuropathic pain. Front. Pain Res. (Lausanne) 2022, 3, 1013577. [Google Scholar] [CrossRef] [PubMed]
  133. Inceoglu, B.; Bettaieb, A.; Trindade da Silva, C.A.; Lee, K.S.; Haj, F.G.; Hammock, B.D. Endoplasmic reticulum stress in the peripheral nervous system is a significant driver of neuropathic pain. Proc. Natl. Acad. Sci. USA 2015, 112, 9082–9087. [Google Scholar] [CrossRef]
  134. Oñate, M.; Court, F.A.; Hetz, C. Bursting the unfolded protein response accelerates axonal regeneration. Neural Regen. Res. 2016, 11, 892–893. [Google Scholar]
  135. Ohtake, Y.; Matsuhisa, K.; Kaneko, M.; Kanemoto, S.; Asada, R.; Imaizumi, K.; Saito, A. Axonal Activation of the Unfolded Protein Response Promotes Axonal Regeneration Following Peripheral Nerve Injury. Neuroscience 2018, 375, 34–48. [Google Scholar] [CrossRef]
  136. Perner, C.; Krüger, E. Endoplasmic Reticulum Stress and Its Role in Homeostasis and Immunity of Central and Peripheral Neurons. Front. Immunol. 2022, 13, 859703. [Google Scholar] [CrossRef] [PubMed]
  137. Meza-Menchaca, T.; Albores-Medina, A.; Heredia-Mendez, A.J.; Ruíz-May, E.; Ricaño-Rodríguez, J.; Gallegos-García, V.; Esquivel, A.; Vettoretti-Maldonado, G.; Campos-Parra, A.D. Revisiting Epigenetics Fundamentals and Its Biomedical Implications. Int. J. Mol. Sci. 2024, 25, 7927. [Google Scholar] [CrossRef] [PubMed]
  138. Shin, J.E.; Cho, Y. Epigenetic Regulation of Axon Regeneration after Neural Injury. Mol. Cells 2017, 40, 10–16. [Google Scholar] [CrossRef]
  139. Zhang, W.; Jiao, B.; Yu, S.; Zhang, C.; Zhang, K.; Liu, B.; Zhang, X. Histone deacetylase as emerging pharmacological therapeutic target for neuropathic pain: From epigenetic to selective drugs. CNS Neurosci. Ther. 2024, 30, e14745. [Google Scholar] [CrossRef]
  140. Gölzenleuchter, M.; Kanwar, R.; Zaibak, M.; Al Saiegh, F.; Hartung, T.; Klukas, J.; Smalley, R.L.; Cunningham, J.M.; Figueroa, M.E.; Schroth, G.P.; et al. Plasticity of DNA methylation in a nerve injury model of pain. Epigenetics 2015, 10, 200–212. [Google Scholar] [CrossRef]
  141. Liang, L.; Gu, X.; Zhao, J.Y.; Wu, S.; Miao, X.; Xiao, J.; Mo, K.; Zhang, J.; Lutz, B.M.; Bekker, A.; et al. G9a participates in nerve injury-induced Kcna2 downregulation in primary sensory neurons. Sci. Rep. 2016, 6, 37704. [Google Scholar] [CrossRef]
  142. Pollema-Mays, S.L.; Centeno, M.V.; Apkarian, A.V.; Martina, M. Expression of DNA methyltransferases in adult dorsal root ganglia is cell-type specific and up regulated in a rodent model of neuropathic pain. Front. Cell Neurosci. 2014, 8, 217. [Google Scholar] [CrossRef] [PubMed]
  143. Chamessian, A.G.; Qadri, Y.J.; Cummins, M.; Hendrickson, M.; Berta, T.; Buchheit, T.; Van de Ven, T. 5-Hydroxymethylcytosine (5hmC) and Ten-eleven translocation 1-3 (TET1-3) proteins in the dorsal root ganglia of mouse: Expression and dynamic regulation in neuropathic pain. Somatosens. Mot. Res. 2017, 34, 72–79. [Google Scholar] [CrossRef]
  144. Nagata, K.; Hama, I.; Kiryu-Seo, S.; Kiyama, H. microRNA-124 is down regulated in nerve-injured motor neurons and it potentially targets mRNAs for KLF6 and STAT3. Neuroscience 2014, 256, 426–432. [Google Scholar] [CrossRef] [PubMed]
  145. Sakai, A.; Suzuki, H. Nerve injury-induced upregulation of miR-21 in the primary sensory neurons contributes to neuropathic pain in rats. Biochem. Biophys. Res. Commun. 2013, 435, 176–181. [Google Scholar] [CrossRef] [PubMed]
  146. Zhao, X.; Tang, Z.; Zhang, H.; Atianjoh, F.E.; Zhao, J.Y.; Liang, L.; Wang, W.; Guan, X.; Kao, S.C.; Tiwari, V.; et al. A long noncoding RNA contributes to neuropathic pain by silencing Kcna2 in primary afferent neurons. Nat. Neurosci. 2013, 16, 1024–1031. [Google Scholar] [CrossRef]
  147. Yang, L.; Lin, C.; Liu, W.; Zhang, J.; Ohgi, K.A.; Grinstein, J.D.; Dorrestein, P.C.; Rosenfeld, M.G. ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell 2011, 147, 773–788. [Google Scholar] [CrossRef]
  148. Pinto-Costa, R.; Sousa, S.C.; Leite, S.C.; Nogueira-Rodrigues, J.; Ferreira da Silva, T.; Machado, D.; Marques, J.; Costa, A.C.; Liz, M.A.; Bartolini, F.; et al. Profilin 1 delivery tunes cytoskeletal dynamics toward CNS axon regeneration. J. Clin. Investig. 2020, 130, 2024–2040. [Google Scholar] [CrossRef]
  149. Hur, E.M.; Saijilafu; Zhou, F.Q. Growing the growth cone: Remodeling the cytoskeleton to promote axon regeneration. Trends Neurosci. 2012, 35, 164–174. [Google Scholar] [CrossRef]
  150. Takei, Y.; Teng, J.; Harada, A.; Hirokawa, N. Defects in axonal elongation and neuronal migration in mice with disrupted tau and map1b genes. J. Cell Biol. 2000, 150, 989–1000. [Google Scholar] [CrossRef]
  151. Watabe-Uchida, M.; Govek, E.E.; Van Aelst, L. Regulators of Rho GTPases in neuronal development. J. Neurosci. 2006, 26, 10633–10635. [Google Scholar] [CrossRef] [PubMed]
  152. Iwata, K.; Shinoda, M. Role of neuron and non-neuronal cell communication in persistent orofacial pain. J. Dent. Anesth. Pain Med. 2019, 19, 77–82. [Google Scholar] [CrossRef] [PubMed]
  153. Jessen, K.R.; Arthur-Farraj, P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019, 67, 421–437. [Google Scholar] [CrossRef]
  154. Avraham, O.; Deng, P.Y.; Jones, S.; Kuruvilla, R.; Semenkovich, C.F.; Klyachko, V.A.; Cavalli, V. Satellite glial cells promote regenerative growth in sensory neurons. Nat. Commun. 2020, 11, 4891. [Google Scholar] [CrossRef]
  155. Parfejevs, V.; Debbache, J.; Shakhova, O.; Schaefer, S.M.; Glausch, M.; Wegner, M.; Suter, U.; Riekstina, U.; Werner, S.; Sommer, L. Injury-activated glial cells promote wound healing of the adult skin in mice. Nat. Commun. 2018, 9, 236. [Google Scholar] [CrossRef] [PubMed]
  156. Hanani, M. How Is Peripheral Injury Signaled to Satellite Glial Cells in Sensory Ganglia? Cells 2022, 11, 512. [Google Scholar] [CrossRef]
  157. Qiu, X.; Yang, Y.; Da, X.; Wang, Y.; Chen, Z.; Xu, C. Satellite glial cells in sensory ganglia play a wider role in chronic pain via multiple mechanisms. Neural Regen. Res. 2024, 19, 1056–1063. [Google Scholar] [CrossRef]
  158. Retamal, M.A.; Riquelme, M.A.; Stehberg, J.; Alcayaga, J. Connexin43 Hemichannels in Satellite Glial Cells, Can They Influence Sensory Neuron Activity? Front. Mol. Neurosci. 2017, 10, 374. [Google Scholar] [CrossRef] [PubMed]
  159. Kaji, K.; Shinoda, M.; Honda, K.; Unno, S.; Shimizu, N.; Iwata, K. Connexin 43 contributes to ectopic orofacial pain following inferior alveolar nerve injury. Mol. Pain 2016, 12, 1744806916633704. [Google Scholar] [CrossRef]
  160. Hanani, M.; Spray, D.C. Emerging importance of satellite glia in nervous system function and dysfunction. Nat. Rev. Neurosci. 2020, 21, 485–498. [Google Scholar] [CrossRef]
  161. Reichling, D.B.; Levine, J.D. Critical role of nociceptor plasticity in chronic pain. Trends Neurosci. 2009, 32, 611–618. [Google Scholar] [CrossRef] [PubMed]
  162. Jang, K.; Garraway, S.M. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. Neurobiol. Pain 2024, 15, 100151. [Google Scholar] [CrossRef] [PubMed]
  163. Renthal, W.; Tochitsky, I.; Yang, L.; Cheng, Y.C.; Li, E.; Kawaguchi, R.; Geschwind, D.H.; Woolf, C.J. Transcriptional Reprogramming of Distinct Peripheral Sensory Neuron Subtypes after Axonal Injury. Neuron 2020, 108, 128–144.e9. [Google Scholar] [CrossRef]
  164. Wang, Y.; Li, W.Y.; Li, Z.G.; Guan, L.X.; Deng, L.X. Transcriptional and Epigenetic Regulation in Injury-Mediated Neuronal Dendritic Plasticity. Neurosci. Bull. 2017, 33, 85–94. [Google Scholar] [CrossRef] [PubMed]
  165. Martin, S.L.; Reid, A.J.; Verkhratsky, A.; Magnaghi, V.; Faroni, A. Gene expression changes in dorsal root ganglia following peripheral nerve injury: Roles in inflammation, cell death and nociception. Neural Regen. Res. 2019, 14, 939–947. [Google Scholar]
  166. Jung, M.; Dourado, M.; Maksymetz, J.; Jacobson, A.; Laufer, B.I.; Baca, M.; Foreman, O.; Hackos, D.H.; Riol-Blanco, L.; Kaminker, J.S. Cross-species transcriptomic atlas of dorsal root ganglia reveals species-specific programs for sensory function. Nat. Commun. 2023, 14, 366. [Google Scholar] [CrossRef]
  167. Hetman, M.; Pietrzak, M. Emerging roles of the neuronal nucleolus. Trends Neurosci. 2012, 35, 305–314. [Google Scholar] [CrossRef]
  168. Hetman, M.; Slomnicki, L.P. Ribosomal biogenesis as an emerging target of neurodevelopmental pathologies. J. Neurochem. 2019, 148, 325–347. [Google Scholar] [CrossRef]
  169. Chen, X.; Tang, S.J. Neural Circuitry Polarization in the Spinal Dorsal Horn (SDH): A Novel Form of Dysregulated Circuitry Plasticity during Pain Pathogenesis. Cells 2024, 13, 398. [Google Scholar] [CrossRef]
  170. Ackery, A.D.; Norenberg, M.D.; Krassioukov, A. Calcitonin gene-related peptide immunoreactivity in chronic human spinal cord injury. Spinal Cord 2007, 45, 678–686. [Google Scholar] [CrossRef]
  171. Li, J.; Baccei, M.L. Neonatal tissue damage facilitates nociceptive synaptic input to the developing superficial dorsal horn via NGF-dependent mechanisms. Pain 2011, 152, 1846–1855. [Google Scholar] [CrossRef] [PubMed]
  172. Terayama, R.; Uchibe, K. Reorganization of synaptic inputs to spinal dorsal horn neurons in neuropathic pain. Int. J. Neurosci. 2022, 132, 1210–1216. [Google Scholar] [CrossRef] [PubMed]
  173. Graham, R.D.; Sankarasubramanian, V.; Lempka, S.F. Dorsal Root Ganglion Stimulation for Chronic Pain: Hypothesized Mechanisms of Action. J. Pain 2022, 23, 196–211. [Google Scholar] [CrossRef] [PubMed]
  174. Hayward, R.; Moore, S.; Artun, D.; Madhavan, A.; Harte, E.; Torres-Pérez, J.V.; Nagy, I. Transcriptional reprogramming post-peripheral nerve injury: A systematic review. Neurobiol. Dis. 2024, 200, 106624. [Google Scholar] [CrossRef]
  175. Chen, L.; Li, Y.; Zhu, L.; Jin, H.; Kang, X.; Feng, Z. Single-cell RNA sequencing in the context of neuropathic pain: Progress, challenges, and prospects. Transl. Res. 2023, 251, 96–103. [Google Scholar] [CrossRef]
  176. Stenz, L.; Carré, J.L.; Luthi, F.; Vuistiner, P.; Burrus, C.; Paoloni-Giacobino, A.; Léger, B. Genome-Wide Epigenomic Analyses in Patients With Nociceptive and Neuropathic Chronic Pain Subtypes Reveals Alterations in Methylation of Genes Involved in the Neuro-Musculoskeletal System. J. Pain 2022, 23, 326–336. [Google Scholar] [CrossRef]
  177. Li, Y.Z.; Ji, R.R. Gene therapy for chronic pain management. Cell Rep. Med. 2024, 5, 101756. [Google Scholar] [CrossRef]
  178. Hung, A.L.; Lim, M.; Doshi, T.L. Targeting cytokines for treatment of neuropathic pain. Scand. J. Pain 2017, 17, 287–293. [Google Scholar] [CrossRef] [PubMed]
  179. Pawlik, K.; Mika, J. Targeting Members of the Chemokine Family as a Novel Approach to Treating Neuropathic Pain. Molecules 2023, 28, 5766. [Google Scholar] [CrossRef]
  180. Ivanova, E.A.; Vasilchuk, A.G.; Voronina, T.A. Protein Kinase A Inhibitor Attenuates the Antinociceptive Effect of NMDA-Receptor Channel Antagonists in the Capsaicin Test in Mice. Bull. Exp. Biol. Med. 2024, 177, 231–234. [Google Scholar] [CrossRef]
  181. Géranton, S.M. Targeting epigenetic mechanisms for pain relief. Curr. Opin. Pharmacol. 2012, 12, 35–41. [Google Scholar] [CrossRef] [PubMed]
  182. Louwies, T.; Ligon, C.O.; Johnson, A.C.; Greenwood-Van Meerveld, B. Targeting epigenetic mechanisms for chronic visceral pain: A valid approach for the development of novel therapeutics. Neurogastroenterol. Motil. 2019, 31, e13500. [Google Scholar] [CrossRef]
  183. Wolfe, D.; Mata, M.; Fink, D.J. Targeted drug delivery to the peripheral nervous system using gene therapy. Neurosci. Lett. 2012, 527, 85–89. [Google Scholar] [CrossRef] [PubMed]
  184. Jayakar, S.; Shim, J.; Jo, S.; Bean, B.P.; Singeç, I.; Woolf, C.J. Developing nociceptor-selective treatments for acute and chronic pain. Sci. Transl. Med. 2021, 13, eabj9837. [Google Scholar] [CrossRef]
  185. Cao, B.; Xu, Q.; Shi, Y.; Zhao, R.; Li, H.; Zheng, J.; Liu, F.; Wan, Y.; Wei, B. Pathology of pain and its implications for therapeutic interventions. Signal. Transduct. Target Ther. 2024, 9, 155. [Google Scholar] [PubMed]
  186. Rijsdijk, M.; Smits, H.M.; Azizoglu, H.R.; Brugman, S.; van de Burgt, Y.; van Charldorp, T.C.; van Gelder, D.J.; de Grauw, J.C.; van Lange, E.A.; Meye, F.J.; et al. Identifying patient subgroups in the heterogeneous chronic pain population using cluster analysis. J. Pain 2025, 28, 104792. [Google Scholar] [CrossRef]
  187. Tracey, I.; Woolf, C.J.; Andrews, N.A. Composite Pain Biomarker Signatures for Objective Assessment and Effective Treatment. Neuron 2019, 101, 783–800. [Google Scholar] [CrossRef]
Figure 1. Nociceptors are specialized sensory neurons that can become activated under pathological conditions in response to a variety of stimuli, including thermal, mechanical, and chemical signals. Under physiological states, nociceptors detect noxious stimuli to initiate protective responses; however, during pathological states such as inflammation or nerve injury, their sensitivity can become markedly enhanced. Abbreviations: IL-1β (Interleukin 1 beta), IL-6 (interleukin-6), TNF-α (tumor necrosis factor alpha), ATP (adenosine triphosphate), GPCRs (G protein-coupled receptors), ASICs (acid-sensing ion channels), P2X3 (purinergic receptor P2X, ligand-gated ion channel 3), Nav1.7 (voltage-gated sodium channel alpha subunit 1.7), Nav1.8 (voltage-gated sodium channel alpha subunit 1.8), and Nav1.9 (voltage-gated sodium channel alpha subunit 1.9).
Figure 1. Nociceptors are specialized sensory neurons that can become activated under pathological conditions in response to a variety of stimuli, including thermal, mechanical, and chemical signals. Under physiological states, nociceptors detect noxious stimuli to initiate protective responses; however, during pathological states such as inflammation or nerve injury, their sensitivity can become markedly enhanced. Abbreviations: IL-1β (Interleukin 1 beta), IL-6 (interleukin-6), TNF-α (tumor necrosis factor alpha), ATP (adenosine triphosphate), GPCRs (G protein-coupled receptors), ASICs (acid-sensing ion channels), P2X3 (purinergic receptor P2X, ligand-gated ion channel 3), Nav1.7 (voltage-gated sodium channel alpha subunit 1.7), Nav1.8 (voltage-gated sodium channel alpha subunit 1.8), and Nav1.9 (voltage-gated sodium channel alpha subunit 1.9).
Biology 14 00788 g001
Figure 2. Schematic representation of the nociceptive transmission pathway. This figure illustrates the key anatomical components involved in the transmission of pain signals, beginning with the activation of peripheral nociceptors and culminating in the perception of pain in the cerebral cortex. It outlines the sequential steps of signal transduction, including primary afferent neuron activation, synaptic transmission in the dorsal horn of the spinal cord, ascending pathways (such as the spinothalamic tract), and the integration of nociceptive signals in the cerebral cortex. The red-colored neuron represents the first-order neuron, the orange one corresponds to the second-order neuron, and the green neuron denotes the third-order neuron.
Figure 2. Schematic representation of the nociceptive transmission pathway. This figure illustrates the key anatomical components involved in the transmission of pain signals, beginning with the activation of peripheral nociceptors and culminating in the perception of pain in the cerebral cortex. It outlines the sequential steps of signal transduction, including primary afferent neuron activation, synaptic transmission in the dorsal horn of the spinal cord, ascending pathways (such as the spinothalamic tract), and the integration of nociceptive signals in the cerebral cortex. The red-colored neuron represents the first-order neuron, the orange one corresponds to the second-order neuron, and the green neuron denotes the third-order neuron.
Biology 14 00788 g002
Figure 3. Graphical representation of genetic and epigenetic modifications in nociceptors induced by injury. Abbreviations: DRG (dorsal root ganglion), DNA (deoxyribonucleic acid), RNA (ribonucleic acid), VGSC (voltage-gated sodium channel), VGCC (voltage-gated calcium channel), TRPV1 (transient receptor potential vanilloid 1), TRPA1 (transient receptor potential ankyrin 1), TRPM8 (transient receptor potential melastatin 8), HAT (histone acetyltransferase), VGKC (voltage-gated potassium channel), and HDAC (histone deacetylase).
Figure 3. Graphical representation of genetic and epigenetic modifications in nociceptors induced by injury. Abbreviations: DRG (dorsal root ganglion), DNA (deoxyribonucleic acid), RNA (ribonucleic acid), VGSC (voltage-gated sodium channel), VGCC (voltage-gated calcium channel), TRPV1 (transient receptor potential vanilloid 1), TRPA1 (transient receptor potential ankyrin 1), TRPM8 (transient receptor potential melastatin 8), HAT (histone acetyltransferase), VGKC (voltage-gated potassium channel), and HDAC (histone deacetylase).
Biology 14 00788 g003
Figure 4. Schematic representation of the regenerative process of myelinated nociceptors. Following injury, resident macrophages release several pro-inflammatory cytokines (e.g., IL-1β, IL-6, and TNF-α), which recruit additional macrophages to the site of damage to promote axonal regeneration. In parallel, Schwann cells contribute to the regenerative environment by secreting neurotrophic factors such as GDNF, NGF, and BDNF, thereby supporting and accelerating the growth of injured axons. Abbreviations: GDNF (glial cell line-derived neurotrophic factor), NGF (nerve growth factor), and BDNF (brain-derived neurotrophic factor).
Figure 4. Schematic representation of the regenerative process of myelinated nociceptors. Following injury, resident macrophages release several pro-inflammatory cytokines (e.g., IL-1β, IL-6, and TNF-α), which recruit additional macrophages to the site of damage to promote axonal regeneration. In parallel, Schwann cells contribute to the regenerative environment by secreting neurotrophic factors such as GDNF, NGF, and BDNF, thereby supporting and accelerating the growth of injured axons. Abbreviations: GDNF (glial cell line-derived neurotrophic factor), NGF (nerve growth factor), and BDNF (brain-derived neurotrophic factor).
Biology 14 00788 g004
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

García-Domínguez, M. Injury-Driven Structural and Molecular Modifications in Nociceptors. Biology 2025, 14, 788. https://doi.org/10.3390/biology14070788

AMA Style

García-Domínguez M. Injury-Driven Structural and Molecular Modifications in Nociceptors. Biology. 2025; 14(7):788. https://doi.org/10.3390/biology14070788

Chicago/Turabian Style

García-Domínguez, Mario. 2025. "Injury-Driven Structural and Molecular Modifications in Nociceptors" Biology 14, no. 7: 788. https://doi.org/10.3390/biology14070788

APA Style

García-Domínguez, M. (2025). Injury-Driven Structural and Molecular Modifications in Nociceptors. Biology, 14(7), 788. https://doi.org/10.3390/biology14070788

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