Abstract
Chronic pain affects up to a third of the population. Ongoing epidemiology studies suggest that the impact of chronic pain on the population is accelerating [1]. While advances have been made in understanding how chronic pain develops, there are still many important mysteries about how acute pain transitions to a chronic state. In this review, I summarize recent developments in the field with a focus on several areas of emerging research that are likely to have an important impact on the field. These include mechanisms of cellular plasticity that drive chronic pain, evidence of pervasive sex differential mechanisms in chronic pain and the profound impact that next generation sequencing technologies are having on this area of research.
Defining the transition from acute to chronic pain
Chronic pain is defined as pain that persists for at least 3 to 6 months [2]. This definition is clinically useful but it does not give any insight into mechanism. It also does not point out when the transition from acute to chronic pain occurs. A reading of the existing clinical literature on the topic suggests that the transition point is presumed to occur around the 3 to 6 month point, however, we will argue in this review that the preclinical literature suggests that the transition point is actually far earlier than the 3 month time point [3]. Given what we now know about the profound cellular plasticity that is brought about by injury that drives sensitization of the nociceptive system in the peripheral (PNS) and central nervous systems (CNS), it is likely that the transition likely occurs at time points near injury and may even occur concurrently with injury.
Why would such a system exist? Responding to injury is fundamentally important for all organisms. Nociceptors, neurons that are tuned to respond to injurious or potentially injurious stimuli, appeared early in nervous system evolution [4]. These neurons are remarkably plastic. They demonstrate the ability to increase their excitability upon sustained activation in every species where studies have been done to examine this form of intrinsic plasticity [4]. This creates a powerful teaching signal that enhances survival [5] while allowing the organism to recover from insult. An important question is whether this plasticity resolves or if its default mode is persistence. If it is indeed the latter, then injury that can cause sensitization of nociceptors may be doomed to cause a transition to a chronic pain state in most cases.
We know, however, that this is not the case. People have remarkably similar injuries everyday wherein only a minority will continue to have pain after the injury resolves [6]. How, then, can we reconcile our clinical observations with what we now understand about the physiology of nociceptor plasticity? The answer likely lies in a second type of plasticity that is driven by pain resolution mechanisms [7]. The pioneering work of Serhan and colleagues revolutionized the way that we think about inflammation from a physiological process that runs its course to one that will persist far beyond its usefulness unless it is actively resoled by a novel class of lipid mediators called resolvins [8]. This idea has now permeated areas of neurobiology. We now also know, for instance, that the forgetting of aversive memories does not happen through the gradual decay of plasticity mechanisms involved in the original learning, but is instead driven by extinction mechanisms that require new learning [9]. With these paradigms in mind, it may be useful to think of a transition from acute to chronic pain as a default mode that can be overridden by pain resolution mechanisms. This idea is now emerging from numerous studies in the field. Here we will argue that this notion has the potential to revolutionize pain medicine, but that discovery in this area will likely be hard fought, largely because we lack understanding of the basics of nociceptive plasticity in females (of all species) [10].
Modeling the acute to chronic pain transition
Defining preclinical models as “chronic” is challenging because researchers rarely study experimental animals that have had pain for more than 1 month, much less 3 or 6. The argument is often made that this is justified based on proportions of life-span to disease duration, but this defies the physics of time. Having said that, there is still a strong argument to be made for studying plasticity mechanisms relatively early after injury because these mechanisms are likely key drivers of the transition to chronic pain.
Inflammation
Two similar models have been extensively studied in the past few years in terms of understanding how inflammation might lead to the transition to chronic pain: hyperalgesic priming [11–13] and latent sensitization [14]. In the hyperalgesic priming model an animal receives a “priming” stimulus, that is usually an inflammagen (e.g. carrageenan) or a defined inflammatory stimulus (e.g. interleukin 6, IL6) and allowed to recover from the initial nociceptive hypersensitivity (usually thermal and mechanical hyperalgesia). The animal is then challenged with an otherwise subthreshold dose of a different mediator that can now induce a long-lasting pain state. This system has been applied to the hindpaw and also to the cranial dura, creating a new model for migraine headache-like pain [15,16]. In the latent sensitization paradigm the animal is similarly primed but the animal is subsequently treated with an antagonist or inverse agonist of a G-protein coupled receptor (GPCR) that then reveals a hypersensitive state that was not previously apparent [17–21].
Both of these models suggest forms of plasticity in the PNS and CNS that are persistent after the original injury, indicating the presence of a transition to a new homeostatic point for the organism. In the case of hyperalgesic priming it is clear that even though nociceptors may not be actively generating signals, they can easily become activated by stimuli which would ordinarily not have an effect. Interfering with specific translation regulation signaling pathways at the time of injury seems to be the most effective way to impede the development of priming [22–27]. However, several recent studies from Levine’s group have demonstrated multiple forms of priming that have very distinct molecular mechanisms [28–30]. If priming occurs in patient populations it may be critical to understand what mechanism the insult engages to reduce the probability of development of chronic pain. In the case of latent sensitization, CNS-driven, GPCR-mediated mechanisms actively suppress signals that can cause nociceptive hypersensitivity to very rapidly re-emerge if these GPCRs are blocked [17–21]. This suppressive mechanism may be one important way that pain hypersensitivity resolves after severe inflammation or surgery. While a first attempt to demonstrate latent sensitization in people was unsuccessful [31], a subsequent trial showed a statistically significant unmasking of latent sensitization upon administration of high-dose naloxone [32,33].
Nerve injury and neuropathic pain
The most widely accepted models of chronic pain fall into the category of neuropathic pain and usually involve traumatic injury to a nerve (e.g. spared nerve injury, SNI), drug-induced neuropathy (e.g. chemotherapy induced painful neuropathy, CIPN) or metabolic disease (diabetic neuropathy). Some of the earliest evidence for an acute to chronic transition came from studies where it was shown that descending modulatory circuits from the periaqueductal gray (PAG) and rostral ventromedial medulla (RVM) are crucial for promoting pain in the first few weeks after injury [34]. More recent studies have demonstrated that rodents which are protected from the development of neuropathic pain after injury show engagement of descending inhibition at the time of injury to suppress later neuropathic pain [35]. This finding provides good evidence for the idea that the acute to chronic pain transition can occur very early after pain begins and that protection from the transition is due to endogenous pain resolution mechanisms. Strikingly, some human imaging studies provide evidence that people who are protected from developing chronic pain may do so through enhanced descending inhibition [36].
The acute to chronic pain transition in neuropathic pain is not entirely driven by central mechanisms, and is a complex interaction of central and peripheral processes with inherited and environmental risks (Figure 1). Recent human studies have demonstrated that blocking peripheral nerves in patients with neuropathic pain leads to almost immediate pain resolution that returns only after the nerve block wears off [37,38]. This, surprisingly, also occurs in patients who have neuropathic pain driven by stroke [39], suggesting that even CNS injuries may ultimately cause nociceptors to become sensitized and fire action potentials ectopically [40]. These studies point out an important point about CNS plasticity in neuropathic pain (which is often referred to as central sensitization [41]): these mechanisms amplify signals via synaptic gain and may not be capable of generating signals on their own. Therefore, they require an afferent signal to amplify and that afferent signal is likely provided by ectopic or evoked activity in peripheral nerves. Importantly, neuropathic pain in animals and humans is accompanied by the generation of ectopic activity in peripheral nerves [42,43].
Figure 1.
Chronic pain is typically the outcome of interaction between complex inherited polygenic risk, sex, environmental factors, developmental and medical history and triggers like insult or injury. Long term outcomes of chronic pain include changes in molecular configuration and profile, persistent immune signaling, changes in electrophysiology and the connectome, and resulting organism level changes in behavior.
Mechanisms of acute to chronic pain transition involve cellular plasticity
The phenotypic changes occurring in nociceptors after injury driving the transition to a chronic pain state require changes in the molecular profile of these cells [44]. Many recent RNA-seq experiments have somewhat surprisingly shown that wholesale transcriptional changes do not seem to occur in DRG neurons and that those that are reliably found after nerve injury are mostly involved in the regeneration response [45,46]. Instead, translation regulation seems to be the key mechanism that nociceptors use to alter their phenotype toward profound hyperexcitability [26,44,46,47]. Two pathways that have recently emerged as critical for translation of new proteins that play a key role in the development of chronic pain are the MNK-eIF4E signaling axis [26,46] and poly-A binding protein (PABP) [48]. Genetic manipulation of MNK-eIF4E signaling renders mice less susceptible to injury-induced nociceptive hypersensitivity and almost completely abolishes hyperalgesic priming [26]. Tomivosertib (formerly known as eFT508) is a clinical candidate drug for cancer that specifically targets MNK [49,50]. This drug reverses mechanical hypersensitivity, ongoing pain and nociceptor ectopic activity in mouse CIPN models (in males and females) suggesting that this drug may be effective for treating chronic neuropathic pain in people [46]. MNK-elF4E signaling regulates translation at the 5’ end of mRNAs while PABP controls translation at the 3’ end. Recent evidence suggests that 3’ end processing of mRNAs is critical for localization of mRNAs in neurons, as well as neuronal plasticity. Blocking PABP function with a novel RNA-mimetic drug reduces development of chronic pain in response to inflammatory mediators and incision in mice [48]. These studies point to two, mechanistically distinct intervention points for preventing the acute to chronic pain transition.
Ion channels have long been considered prime targets for the treatment of chronic pain. One of the most important in this class is Nav1.7, where the rationale for targeting this protein is clear based on the human genetics [51], although complicated somewhat by recent evidence of anatomical changes brought about by Nav1.7 loss of function mutation [52]. The studies that built this rationale largely identified mutations that led to profound gain or loss of function phenotypes. Interestingly, other, rare mutations have been discovered that lead to enhanced susceptibility of developing a chronic pain state in the presence of other variables, like diabetes and diabetic neuropathic pain [53]. This study establishes a paradigm wherein the interaction of genetics with environment may be a critical factor in determining who will transition to chronic pain states. As DNA sequencing continues to advance into the clinical space, new discoveries along these lines will unquestionably be made. An excellent example of this is the case of two related people with gain of function mutations in Nav1.7 wherein one had a mild pain phenotype and the other severe pain. Whole exome sequencing revealed that a point mutation in Kv7.2 causing a gain of function decreased excitability in induced pluripotent stem cells (iPSCs) -derived sensory neurons from the person with less pain [54]. This gene-gene interaction illustrates the way that modern genetic sequencing technologies can be combined with the utility of iPSC-derived sensory neurons to make fundamental discoveries about pain phenotypes found in people. This study also highlights the key role that voltage gated potassium channels (VGKCs) play in the development of chronic pain. These channels show decreased expression after nerve injury, which results in enhanced excitability [55]. Many of these channels are downregulated at the transcriptional level via an epigenetic mechanism involving G9a [56].
Central circuits play a key role in regulating the transition from acute to chronic pain. Plasticity mechanisms in the spinal dorsal horn involving long term potentiation (LTP) and/or loss of GABAergic efficacy have long been recognized as key factors. The past several years have seen rich gains in our understanding of dorsal horn circuitry but much of this work is still focusing on acute pain, or ablation of circuits prior to injury [57]. It will be interesting to see if different circuits contribute to acute versus chronic pain. An example where this has already been shown is the descending dopaminergic circuit from the A11 nucleus [58]. This circuit inhibits acute and neuropathic pain through a D2-like-mediated mechanism [59]. In hyperalgesic priming models, once the primed state is established, the circuit plays a key role in maintaining priming via D1 and D5 dopamine receptors. Interestingly, this circuit relies on D5 receptors in male mice and D1 receptors in female mice [60]. This brings us to one of the strongest recent themes in pain research: sex differences.
Mechanisms of acute to chronic pain transition are sex differential
Sex differences in basic pain mechanisms is one of the most rapidly expanding areas of discovery in the field (Figure 2) [61]. Most of this experimentation has highlighted sex differences in how pain becomes chronic. The most widely studied of these differences is the role of glia and immune cells in chronic neuropathic pain. It is now clear that there are major differences in the role of astrocytes and microglia wherein certain signaling pathways in these cells play a strong role in promoting pain in male but not female rodents [61–66]. A very recent paper from the Salter lab suggests that this difference boils down to sex differences downstream of the P2X4 receptor including p38 MAPK and IRF5-mediated transcription [67]. Many studies have suggested that this sex difference is driven by sex hormones [65]. However, injury to neonatal mice that drives important changes in adult pain behavior is also sex differential with microglial mechanisms playing a key role in male mice [66]. An outstanding question to be addressed is whether microglia play a role in chronic pain in females but simply use different mediators. In support of this conclusion, most of the studies cited above have observed robust microglial “activation” in females in chronic pain models [65,68]. A similar literature is emerging in our understanding of macrophage (these cells share a common developmental lineage with microglia) contributions to chronic pain in rodents [69]. It may be the case that some mechanisms are sex-specific while others, like redox signaling, can be engaged in males and females [70].
Figure 2.
Sex differences in chronic pain can be envisioned as being built upon the building blocks of baseline differences in the nervous and immune systems, with sex differences becoming more pronounced as a result of sex differential risks (including risk of diseases that share co-morbidity with chronic pain), and sex differences in molecular regulation.
Are there immune cells that promote transition to chronic pain specifically in females? Some studies suggest that T-cells may serve such a function [65], but much less is known about signaling mechanisms through which this occurs. This is also controversial because while some studies have shown that T-cells promote chronic pain, others have shown that T-cells can play a critical role in pain resolution [71,72]. Finally, recent work from the Mogil lab demonstrates that T-cells regulate opioid analgesia in male and female rodents but that this effect is stronger in females [73]. The emerging picture from all of these studies is that T-cells are key mediators of pain plasticity in females but we are just scratching the surface in terms of our understanding of how these cells interact with the nervous system to promote or inhibit chronic pain.
While a great deal of recent work has focused on immune cells and sex differences, many differences in the nervous system have also been discovered. An example in the PNS is that activity-dependent slowing of C-fiber action potentials is more pronounced in female rats than males. This effect is substantially decreased with inflammation, contributing to greater hyperalgesia in females compared to males [74]. Baseline gene expression in human tibial nerves also show sex differential gene expression in several genes known to be neuronally expressed, raising the possibility of differential axonal mRNA transport between sexes [75]. In the CNS, sex differences in neuronal plasticity mechanisms are emerging. We recently showed that neuropathic pain causes shortening of axon initial segments in prefrontal cortex neurons of male but not female mice [76]. This structural change drives cognitive dysfunction that is far more pronounced in male than female mice. Synaptic plasticity mechanisms in pain plasticity also show sex dimorphisms. Although controversial, atypical PKCs (aPKC), in particular PKMζ, have been described as mediators of maintenance of LTP at many CNS synapses [77]. Several previous studies, all conducted exclusively in male mice and rats, suggested that spinal aPKCs are required for the transition from acute to chronic pain [78,79]. More recent studies have demonstrated that this spinal effect is specific for male mice. Removal of the gene encoding PKMζ or applying aPKC blockers to the spinal cord inhibit or reverse, respectively, chronic pain in male but not female mice [80]. Spinal inhibition of aPKC also reverses a stress-mediated pain hypersensitivity caused by placing mice in an environment where they previously received a tonic pain stimulus. Strikingly, this effect only occurs in male mice and can be replicated in male, but not female, human volunteers in an experimental setting [81].
A major gap in knowledge is the extent to which these sex differences will be represented in human populations. The latter study [81] suggests the possibility that many of these differences will translate to humans. If this turns out to be the case, sex differences in chronic pain mechanisms will have a profound impact on therapeutic development, clinical trial design and clinical care.
The RNA sequencing (RNA-seq) revolution
RNA-seq is now widely accessible to most laboratories, allowing unbiased profiling of cellular or tissue transcriptomes in normal tissues and/or in disease states. In the pain field, comprehensive resources are now available that have profiled mouse DRG neurons at the single cell level [82–85] and similar resources are published for many other cell types in the periphery and in the CNS [86,87]. Human DRG [42,88] and trigeminal ganglion (TG) transcriptomes [89,90] have been published allowing for rapid comparison of RNA expression levels between preclinical models and people. A rapidly growing number of transcriptomic studies have been published at various times after nerve injury, or in other neuropathic pain models [91]. Most of these studies have failed to find major transcriptome differences after nerve injury, although some of these studies have detected sex differences that may be related to altered immune cell infiltration [45]. A possible explanation for the relatively small number of transcriptomic changes found in these studies is that single cell sequencing technologies need to be applied to detect transcriptomic changes in neuronal populations brought about by neuropathic pain, with one promising study already performed on a rodent injury model [84]. Improvement of sensitivity is also the focus of research efforts in computational genomics, based on frequentist approaches for large scale testing of correlated hypotheses [92], and Bayesian approaches to identifying gene expression changes in unlabeled single cell samples [93]. A final possibility is that profiling techniques that assess translation of mRNAs, like the cell-type specific translating ribosome affinity purification (TRAP) or ribosome profiling technologies, can provide greater insight into changes in gene expression brought about by injury. In support of this idea, two recent studies using these techniques demonstrate widespread changes in gene expression at the translational level that are not reflected in bulk transcriptomes [46,94]. The application of TRAP sequencing to a CIPN model reveals that many previously described CIPN targets are regulated at the translational level. This study also demonstrates that MNK-eIF4E signaling is a key driver of chronic pain caused by chemotherapy in male and female mice [46].
Many groups are now using RNA-seq to gain insight into pain mechanisms in patient populations. We have recently done this on patients with neuropathic pain who are having their DRGs removed during vertebrectomy. Approximately 20% of DRG neurons taken from patients with neuropathic pain show ectopic activity, drawing a striking parallel to findings from preclinical models [42]. Moreover, changes in transcriptomes are readily observed between chronic pain and non-pain samples. In male patients these transcriptomic changes seem to reflect infiltration and/or phenotypic changes in macrophages that may drive chronic pain. In women transcriptomic changes in pain samples did not parallel findings in men, and may reflect altered gene expression in neurons rather than immune cells [42]. These findings, while not precisely paralleling findings from preclinical models, suggest conservation of some core mechanisms of acute to chronic pain transition discussed above in patients with neuropathic pain. Importantly, the unprecedented public availability of these RNA-seq datasets creates rich possibilities for data-mining projects aimed at discovery of new pain mechanisms and targets.
Therapeutics targeting transition resolution mechanisms
An emerging trend in the study of the acute to chronic pain transition is the concept of pain resolution mechanisms [95]. Initial work in this area focused on interleukin 10 (IL10) and resolvins. While there is compelling evidence for both of these mechanisms, the case for IL10 as an endogenous chronic pain resolution mechanism had progressed substantially recently. Several groundbreaking studies have demonstrated that IL10 from T-cells is a key driver of neuropathic pain resolution [71,72]. IL10 also likely plays a role in resolution of other types of pain, like inflammatory pain. Therapeutic approaches employing this mechanism are under clinical development. They include IL10 gene therapy approaches [96] and IL10-IL4 synerkine [97] as a biologic to accelerate pain resolution.
Exercise has long been recognized as one of the most effective ways to reduce chronic pain. Mechanisms involved in this effect have recently been described and also involve IL10. A history of previous exercise can prevent development of chronic muscle pain brought about by mimicking repeated ischemic insults. This occurs via anti-inflammatory macrophages that increase IL10 synthesis and is effective in males and females [98]. Previous exercise history also enhances resolution of neuropathic pain and this effect is likewise driven by an IL10-dependent mechanism [99]. These and other studies make the strong case for IL10 as a key chronic pain resolution mechanism that is apparently effective in male and female rodents.
Putting scientific advances into action
Remarkable gains are happening in our understanding of how acute pain becomes chronic. These advances in basic science need to begin to influence new ways of thinking about developing drugs for and treating chronic pain. First, the temporal definition of chronic pain, while convenient, is outdated. An operational definition of chronic pain would benefit patients. Second, it is time to recognize that sex differences in chronic pain mechanisms are pervasive and that we have little understanding of how chronic pain develops in females. This is particularly unfortunate given that many forms of chronic pain disproportionately affect females [1]. Preclinical studies that focus on male animals out of convenience rather than scientific rationale should be recognized as incomplete [100]. Finally, it is time that we start implementing what we have learned into drug development and clinical trial design. The FDA has long recognized that biological sex differences may guide decisions about the course of drug testing and approval. As a field we should focus on how mechanisms of acute to chronic pain transition can improve the probability that new drugs for this terrible disease will be approved.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declarations of Interest: None
The authors (Theodore J Price and Pradipta Ray) declare that we have no conflicts of interest for this article.
References Cited
- 1.Nahin RL, Sayer B, Stussman BJ, Feinberg TM: Eighteen-Year Trends in the Prevalence of, and Health Care Use for, Noncancer Pain in the United States: Data from the Medical Expenditure Panel Survey. J Pain 2019. [DOI] [PubMed] [Google Scholar]
- 2.Merskey H (Ed): Classification of chronic pain: Descriptions of chronic pain syndromes and definitions of pain terms: International Association for Study of Pain Press; 1986. [PubMed] [Google Scholar]
- 3.Price TJ, Basbaum AI, Bresnahan J, Chambers JF, De Koninck Y, Edwards RR, Ji RR, Katz J, Kavelaars A, Levine JD, et al. : Transition to chronic pain: opportunities for novel therapeutics. Nat Rev Neurosci 2018, 19:383–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Walters ET: Nociceptive Biology of Molluscs and Arthropods: Evolutionary Clues About Functions and Mechanisms Potentially Related to Pain. Front Physiol 2018, 9:1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Crook RJ, Dickson K, Hanlon RT, Walters ET: Nociceptive sensitization reduces predation risk. Current Biology 2014, 24:1121–1125. [DOI] [PubMed] [Google Scholar]
- 6.Kehlet H, Jensen TS, Woolf CJ: Persistent postsurgical pain: risk factors and prevention. Lancet 2006, 367:1618–1625. [DOI] [PubMed] [Google Scholar]
- 7.Ji RR, Xu ZZ, Strichartz G, Serhan CN: Emerging roles of resolvins in the resolution of inflammation and pain. Trends Neurosci 2011, 34:599–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Serhan CN: Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510:92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Marek R, Sun Y, Sah P: Neural circuits for a top-down control of fear and extinction. Psychopharmacology (Berl) 2019, 236:313–320. [DOI] [PubMed] [Google Scholar]
- 10.Klein SL, Schiebinger L, Stefanick ML, Cahill L, Danska J, de Vries GJ, Kibbe MR, McCarthy MM, Mogil JS, Woodruff TK, et al. : Opinion: Sex inclusion in basic research drives discovery. Proc Natl Acad Sci U S A 2015, 112:5257–5258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Reichling DB, Levine JD: Critical role of nociceptor plasticity in chronic pain. Trends Neurosci 2009, 32:611–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kandasamy R, Price TJ: The pharmacology of nociceptor priming. Handb Exp Pharmacol 2015, 227:15–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Price TJ, Inyang KE: Commonalities between pain and memory mechanisms and their meaning for understanding chronic pain. Prog Mol Biol Transl Sci 2015, 131:409–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Taylor BK, Corder G: Endogenous analgesia, dependence, and latent pain sensitization. Curr Top Behav Neurosci 2014, 20:283–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Burgos-Vega CC, Quigley LD, Avona A, Price T, Dussor G: Dural stimulation in rats causes brain-derived neurotrophic factor-dependent priming to subthreshold stimuli including a migraine trigger. Pain 2016, 157:2722–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Burgos-Vega CC, Quigley LD, Trevisan Dos Santos G, Yan F, Asiedu M, Jacobs B, Motina M, Safdar N, Yousuf H, Avona A, et al. : Non-invasive dural stimulation in mice: A novel preclinical model of migraine. Cephalalgia 2018:333102418779557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Corder G, Doolen S, Donahue RR, Winter MK, Jutras BL, He Y, Hu X, Wieskopf JS, Mogil JS, Storm DR, et al. : Constitutive mu-opioid receptor activity leads to long-term endogenous analgesia and dependence. Science 2013, 341:1394–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Walwyn WM, Chen W, Kim H, Minasyan A, Ennes HS, McRoberts JA, Marvizon JC: Sustained Suppression of Hyperalgesia during Latent Sensitization by mu-, delta-, and kappa-opioid receptors and alpha2A Adrenergic Receptors: Role of Constitutive Activity. J Neurosci 2016, 36:204–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen W, Tache Y, Marvizon JC: Corticotropin-Releasing Factor in the Brain and Blocking Spinal Descending Signals Induce Hyperalgesia in the Latent Sensitization Model of Chronic Pain. Neuroscience 2018, 381:149–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Severino A, Chen W, Hakimian JK, Kieffer BL, Gaveriaux-Ruff C, Walwyn W, Marvizon JCG: Muopioid receptors in nociceptive afferents produce a sustained suppression of hyperalgesia in chronic pain. Pain 2018, 159:1607–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Taylor BK, Sinha GP, Donahue RR, Grachen CM, Moron JA, Doolen S: Opioid receptors inhibit the spinal AMPA receptor Ca(2+) permeability that mediates latent pain sensitization. Exp Neurol 2019, 314:58–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ferrari LF, Bogen O, Chu C, Levine JD: Peripheral administration of translation inhibitors reverses increased hyperalgesia in a model of chronic pain in the rat. J Pain 2013, 14:731–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Melemedjian OK, Tillu DV, Moy JK, Asiedu MN, Mandell EK, Ghosh S, Dussor G, Price TJ: Local translation and retrograde axonal transport of CREB regulates IL-6-induced nociceptive plasticity. Mol Pain 2014, 10:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ferrari LF, Araldi D, Levine JD: Distinct terminal and cell body mechanisms in the nociceptor mediate hyperalgesic priming. J Neurosci 2015, 35:6107–6116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ferrari LF, Bogen O, Reichling DB, Levine JD: Accounting for the delay in the transition from acute to chronic pain: axonal and nuclear mechanisms. J Neurosci 2015, 35:495–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Moy JK, Khoutorsky A, Asiedu MN, Black BJ, Kuhn JL, Barragan-Iglesias P, Megat S, Burton MD, Burgos-Vega CC, Melemedjian OK, et al. : The MNK-eIF4E Signaling Axis Contributes to Injury-Induced Nociceptive Plasticity and the Development of Chronic Pain. J Neurosci 2017, 37:7481–7499. [DOI] [PMC free article] [PubMed] [Google Scholar]; Translation control has emerged as an important theme in the transition to chronic pain. This work describes how phosphorylation of a single residue on an RNA binding protein can control chronic pain development in response to a wide variety of inflammatory mediators.
- 27.Moy JK, Khoutorsky A, Asiedu MN, Dussor G, Price TJ: eIF4E Phosphorylation Influences Bdnf mRNA Translation in Mouse Dorsal Root Ganglion Neurons. Front Cell Neurosci 2018, 12:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Araldi D, Ferrari LF, Levine JD: Repeated Mu-Opioid Exposure Induces a Novel Form of the Hyperalgesic Priming Model for Transition to Chronic Pain. J Neurosci 2015, 35:12502–12517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Araldi D, Ferrari LF, Levine JD: Adenosine-A1 receptor agonist induced hyperalgesic priming type II. Pain 2016, 157:698–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Araldi D, Ferrari LF, Levine JD: Mu-opioid Receptor (MOR) Biased Agonists Induce Biphasic Dose-dependent Hyperalgesia and Analgesia, and Hyperalgesic Priming in the Rat. Neuroscience 2018, 394:60–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pereira MP, Werner MU, Ringsted TK, Rowbotham MC, Taylor BK, Dahl JB: Does naloxone reinstate secondary hyperalgesia in humans after resolution of a burn injury? A placebo-controlled, double-blind, randomized, cross-over study. PLoS One 2013, 8:e64608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pereira MP, Donahue RR, Dahl JB, Werner M, Taylor BK, Werner MU: Endogenous Opioid-Masked Latent Pain Sensitization: Studies from Mouse to Human. PLoS One 2015, 10:e0134441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Springborg AD, Jensen EK, Taylor BK, Werner MU: Effects of target-controlled infusion of high-dose naloxone on pain and hyperalgesia in a human thermal injury model: a study protocol: A randomized, double-blind, placebo-controlled, crossover trial with an enriched design. Medicine (Baltimore) 2016, 95:e5336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ossipov MH, Dussor GO, Porreca F: Central modulation of pain. J Clin Invest 2010, 120:3779–3787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.De Felice M, Sanoja R, Wang R, Vera-Portocarrero L, Oyarzo J, King T, Ossipov MH, Vanderah TW, Lai J, Dussor GO, et al. : Engagement of descending inhibition from the rostral ventromedial medulla protects against chronic neuropathic pain. Pain 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Denk F, McMahon SB, Tracey I: Pain vulnerability: a neurobiological perspective. Nat Neurosci 2014, 17:192–200. [DOI] [PubMed] [Google Scholar]
- 37.Vaso A, Adahan HM, Gjika A, Zahaj S, Zhurda T, Vyshka G, Devor M: Peripheral nervous system origin of phantom limb pain. Pain 2014, 155:1384–1391. [DOI] [PubMed] [Google Scholar]
- 38.Haroutounian S, Nikolajsen L, Bendtsen TF, Finnerup NB, Kristensen AD, Hasselstrom JB, Jensen TS: Primary afferent input critical for maintaining spontaneous pain in peripheral neuropathy. Pain 2014, 155:1272–1279. [DOI] [PubMed] [Google Scholar]
- 39.Haroutounian S, Ford AL, Frey K, Nikolajsen L, Finnerup NB, Neiner A, Kharasch ED, Karlsson P, Bottros MM: How central is central poststroke pain? The role of afferent input in poststroke neuropathic pain: a prospective, open-label pilot study. Pain 2018. [DOI] [PubMed] [Google Scholar]
- 40.Yang Q, Wu Z, Hadden JK, Odem MA, Zuo Y, Crook RJ, Frost JA, Walters ET: Persistent pain after spinal cord injury is maintained by primary afferent activity. J Neurosci 2014, 34:10765–10769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Latremoliere A, Woolf CJ: Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain 2009, 10:895–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.North R, Yan L, Ray P, Rhines L, Tatsui C, Rao G, Johansson C, Kim YH, Zhang B, Dussor G, et al. : Electrophysiologic and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain 2019:in press. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the first paper to comprehensively catalog transcriptomic changes in the DRG of men and women with neuropathic pain. The work shows that ectopic activity develops in DRG neurons from people with neuropathic pain and that transcriptomic changes are frequently sex differential.
- 43.Ratte S, Prescott SA: Afferent hyperexcitability in neuropathic pain and the inconvenient truth about its degeneracy. Curr Opin Neurobiol 2016, 36:31–37. [DOI] [PubMed] [Google Scholar]
- 44.Khoutorsky A, Price TJ: Translational Control Mechanisms in Persistent Pain. Trends Neurosci 2018, 41:100–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lopes DM, Malek N, Edye M, Jager SB, McMurray S, McMahon SB, Denk F: Sex differences in peripheral not central immune responses to pain-inducing injury. Sci Rep 2017, 7:16460. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper highlights important sex differences in the transcriptome of DRG after nerve injury with the surprising finding that alterations in the CNS are relatively sparse
- 46.Megat S, Ray PR, Moy JK, Lou TF, Barragan-Iglesias P, Li Y, Pradhan G, Wanghzou A, Ahmad A, Burton MD, et al. : Nociceptor Translational Profiling Reveals the Ragulator-Rag GTPase Complex as a Critical Generator of Neuropathic Pain. J Neurosci 2019, 39:393–411. [DOI] [PMC free article] [PubMed] [Google Scholar]; This work identifies mTORC1 and eIF pathways as gateways to translational control in a mouse model of chemotherapy induced pain. TRAP-seq based whole transcriptome assays provide a clear picture of nascent translation, and potential mechanisms at work.
- 47.Barragan-Iglesias P, Kuhn J, Vidal-Cantu GC, Salinas-Abarca AB, Granados-Soto V, Dussor GO, Campbell ZT, Price TJ: Activation of the integrated stress response in nociceptors drives methylglyoxal-induced pain. Pain 2019, 160:160–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Barragan-Iglesias P, Lou TF, Bhat VD, Megat S, Burton MD, Price TJ, Campbell ZT: Inhibition of Poly(A)-binding protein with a synthetic RNA mimic reduces pain sensitization in mice. Nat Commun 2018, 9:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Reich SH, Sprengeler PA, Chiang GG, Appleman JR, Chen J, Clarine J, Eam B, Ernst JT, Han Q, Goel VK, et al. : Structure-based Design of Pyridone-aminal eFT508 Targeting Dysregulated Translation by Selective Mitogen-activated Protein Kinase Interacting Kinases 1 and 2 (MNK½) Inhibition. J Med Chem 2018. [DOI] [PubMed] [Google Scholar]
- 50.Stumpf CR, Chen J, Goel VK, Parker GS, Thompson PA, Webster KR: Inhibition of MNK by eFT508 reprograms T-cell signaling to promote an anti-tumor immune response. In American Association for Cancer Research; Edited by. Chicago, IL; 2018. [Google Scholar]
- 51.Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD: The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019, 99:1079–1151. [DOI] [PubMed] [Google Scholar]
- 52.McDermott LA, Weir GA, Themistocleous AC, Segerdahl AR, Blesneac I, Baskozos G, Clark AJ, Millar V, Peck LJ, Ebner D, et al. : Defining the Functional Role of NaV1.7 in Human Nociception. Neuron 2019:in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Blesneac I, Themistocleous AC, Fratter C, Conrad LJ, Ramirez JD, Cox JJ, Tesfaye S, Shillo PR, Rice ASC, Tucker SJ, et al. : Rare NaV1.7 variants associated with painful diabetic peripheral neuropathy. Pain 2018, 159:469–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mis MA, Yang Y, Tanaka BS, Gomis-Perez C, Liu S, Dib-Hajj F, Adi T, Garcia-Milian R, Schulman BR, Dib-Hajj SD, et al. : Resilience to Pain: A Peripheral Component Identified Using Induced Pluripotent Stem Cells and Dynamic Clamp. J Neurosci 2019, 39:382–392. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper is remarkable because it demonstrates how two people with gain of function mutations in Nav1.7 can have wildly divergent pain phenotypes. The difference boils down to another mutation in a key VGKC. The work is an elegant demonstration of how patient-derived iPSCs can be used to achieve astonishing molecular insight.
- 55.Simms BA, Zamponi GW: Neuronal voltage-gated calcium channels: structure, function, and dysfunction. Neuron 2014, 82:24–45. [DOI] [PubMed] [Google Scholar]
- 56.Laumet G, Garriga J, Chen SR, Zhang Y, Li DP, Smith TM, Dong Y, Jelinek J, Cesaroni M, Issa JP, et al. : G9a is essential for epigenetic silencing of K(+) channel genes in acute-to-chronic pain transition. Nat Neurosci 2015, 18:1746–1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Peirs C, Seal RP: Neural circuits for pain: Recent advances and current views. Science 2016, 354:578–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kim JY, Tillu DV, Quinn TL, Mejia GL, Shy A, Asiedu MN, Murad E, Schumann AP, Totsch SK, Sorge RE, et al. : Spinal dopaminergic projections control the transition to pathological pain plasticity via a D1/D5-mediated mechanism. J Neurosci 2015, 35:6307–6317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Liu S, Tang Y, Shu H, Tatum D, Bai Q, Crawford J, Xing Y, Lobo MK, Bellinger L, Kramer P, et al. : Dopamine receptor D2, but not D1, mediates descending dopaminergic pathway-produced analgesic effect in a trigeminal neuropathic pain mouse model. Pain 2019, 160:334–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Megat S, Shiers S, Moy JK, Barragan-Iglesias P, Pradhan G, Seal RP, Dussor G, Price TJ: A Critical Role for Dopamine D5 Receptors in Pain Chronicity in Male Mice. J Neurosci 2018, 38:379–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rosen S, Ham B, Mogil JS: Sex differences in neuroimmunity and pain. J Neurosci Res 2017, 95:500–508. [DOI] [PubMed] [Google Scholar]
- 62.Paige C, Maruthy GB, Mejia G, Dussor G, Price T: Spinal Inhibition of P2XR or p38 Signaling Disrupts Hyperalgesic Priming in Male, but not Female, Mice. Neuroscience 2018, 385:133–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Taves S, Berta T, Liu DL, Gan S, Chen G, Kim YH, Van de Ven T, Laufer S, Ji RR: Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: Sex-dependent microglial signaling in the spinal cord. Brain Behav Immun 2016, 55:70–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gutierrez S, Hayashida K, Eisenach JC: The puerperium alters spinal cord plasticity following peripheral nerve injury. Neuroscience 2013, 228:301–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sorge RE, Mapplebeck JC, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin JS, Sotocinal SG, Chen D, et al. : Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci 2015, 18:1081–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Moriarty O, YuShan T, Sengar A, Salter MW, Beggs S, Walker S: Priming of adult incision response by early life injury: neonatal microglial inhibition has persistent but sexually dimorphic effects in adult rats. The Journal of neuroscience : the official journal of the Society for Neuroscience 2019:in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Mapplebeck JCS, Dalgarno R, Tu Y, Moriarty O, Beggs S, Kwok CHT, Halievski K, Assi S, Mogil JS, Trang T, et al. : Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats. Pain 2018, 159:1752–1763. [DOI] [PubMed] [Google Scholar]; This manuscript demonstrates that the sex difference in microglial promotion of chronic pain depends on the P2X4 receptor. It also shows that differences observed in mice are conserved in rats.
- 68.Inyang KE, Szabo-Pardi T, Wentworth E, McDougal TA, Dussor G, Burton MD, Price TJ: The antidiabetic drug metformin prevents and reverses neuropathic pain and spinal cord microglial activation in male but not female mice. Pharmacol Res 2018, 139:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Song Z, Xie W, Strong JA, Berta T, Ulrich-Lai YM, Guo Q, Zhang JM: High-fat diet exacerbates postoperative pain and inflammation in a sex-dependent manner. Pain 2018, 159:1731–1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Shepherd AJ, Copits BA, Mickle AD, Karlsson P, Kadunganattil S, Haroutounian S, Tadinada SM, de Kloet AD, Valtcheva MV, McIlvried LA, et al. : Angiotensin II Triggers Peripheral Macrophage-to-Sensory Neuron Redox Crosstalk to Elicit Pain. J Neurosci 2018, 38:7032–7057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Krukowski K, Eijkelkamp N, Laumet G, Hack CE, Li Y, Dougherty PM, Heijnen CJ, Kavelaars A: CD8+ T Cells and Endogenous IL-10 Are Required for Resolution of Chemotherapy-Induced Neuropathic Pain. J Neurosci 2016, 36:11074–11083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Laumet G, Edralin JD, Dantzer R, Heijnen CJ, Kavelaars A: Cisplatin educates CD8+ T cells to prevent and resolve chemotherapy-induced peripheral neuropathy in mice. Pain 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rosen SF, Ham B, Haichin M, Walters IC, Tohyama S, Sotocinal SG, Mogil JS: Increased pain sensitivity and decreased opioid analgesia in T-cell-deficient mice and implications for sex differences. Pain 2019, 160:358–366. [DOI] [PubMed] [Google Scholar]
- 74.Dickie AC, McCormick B, Lukito V, Wilson KL, Torsney C: Inflammatory Pain Reduces C Fiber Activity-Dependent Slowing in a Sex-Dependent Manner, Amplifying Nociceptive Input to the Spinal Cord. J Neurosci 2017, 37:6488–6502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ray P, Khan J, Wangzhou A, Tavares-Ferreira D, Akopian AN, Dussor G, Price TJ: Transcriptome analysis of the human tibial nerve identifies sexually dimorphic expression of genes involved in pain, inflammation and neuro-immunity. Frontiers in molecular neuroscience 2019:in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Shiers S, Pradhan G, Mwirigi J, Mejia G, Ahmad A, Kroener S, Price T: Neuropathic Pain Creates an Enduring Prefrontal Cortex Dysfunction Corrected by the Type II Diabetic Drug Metformin But Not by Gabapentin. J Neurosci 2018, 38:7337–7350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sacktor TC, Hell JW: The genetics of PKMzeta and memory maintenance. Sci Signal 2017, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Laferriere A, Pitcher MH, Haldane A, Huang Y, Cornea V, Kumar N, Sacktor TC, Cervero F, Coderre TJ: PKMzeta is essential for spinal plasticity underlying the maintenance of persistent pain. Mol Pain 2011, 7:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Asiedu M, Ossipov MH, Kaila K, Price TJ: Acetazolamide and midazolam act synergistically to inhibit neuropathic pain. Pain 2010, 148:302–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Nasir H, Mahboubi H, Gyawali S, Ding S, Mickeviciute A, Ragavendran JV, Laferriere A, Stochaj U, Coderre TJ: Consistent sex-dependent effects of PKMzeta gene ablation and pharmacological inhibition on the maintenance of referred pain. Mol Pain 2016, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Martin LJ, Acland EL, Cho C, Gandhi W, Chen D, Corley E, Kadoura B, Levy T, Mirali S, Tohyama S, et al. : Male-Specific Conditioned Pain Hypersensitivity in Mice and Humans. Curr Biol 2019, 29:192–201 e194. [DOI] [PubMed] [Google Scholar]
- 82.Hockley JRF, Taylor TS, Callejo G, Wilbrey AL, Gutteridge A, Bach K, Winchester WJ, Bulmer DC, McMurray G, Smith ESJ: Single-cell RNAseq reveals seven classes of colonic sensory neuron. Gut 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]; While several single cell RNA-seq papers on DRG have been published, this is the first to do so on a specific cell population. The work has important implications for studying and understanding visceral pain.
- 83.Usoskin D, Furlan A, Islam S, Abdo H, Lonnerberg P, Lou D, Hjerling-Leffler J, Haeggstrom J, Kharchenko O, Kharchenko PV, et al. : Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat Neurosci 2015, 18:145–153. [DOI] [PubMed] [Google Scholar]
- 84.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. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the only paper to date that shows different responses to injury in different sensory neuronal subpopulations at single cell resolution in a mouse model.
- 85.Li CL, Li KC, Wu D, Chen Y, Luo H, Zhao JR, Wang SS, Sun MM, Lu YJ, Zhong YQ, et al. : Somatosensory neuron types identified by high-coverage single-cell RNA-sequencing and functional heterogeneity. Cell Res 2016, 26:83–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tabula Muris C, Overall c, Logistical c, Organ c processing, Library p sequencing, Computational data a, Cell type a, Writing g, et al. : Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 2018, 562:367–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zeisel A, Hochgerner H, Lonnerberg P, Johnsson A, Memic F, van der Zwan J, Haring M, Braun E, Borm LE, La Manno G, et al. : Molecular Architecture of the Mouse Nervous System. Cell 2018, 174:999–1014.e1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ray P, Torck A, Quigley L, Wangzhou A, Neiman M, Rao C, Lam T, Kim JY, Kim TH, Zhang MQ, et al. : Comparative transcriptome profiling of the human and mouse dorsal root ganglia: an RNA-seq-based resource for pain and sensory neuroscience research. Pain 2018, 159:1325–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Flegel C, Schobel N, Altmuller J, Becker C, Tannapfel A, Hatt H, Gisselmann G: RNA-Seq Analysis of Human Trigeminal and Dorsal Root Ganglia with a Focus on Chemoreceptors. PLoS One 2015, 10:e0128951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.LaPaglia DM, Sapio MR, Burbelo PD, Thierry-Mieg J, Thierry-Mieg D, Raithel SJ, Ramsden CE, Iadarola MJ, Mannes AJ: RNA-Seq investigations of human post-mortem trigeminal ganglia. Cephalalgia 2018, 38:912–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Starobova H, WAH S, Lewis RJ, Vetter I: Transcriptomics in pain research: insights from new and old technologies. Mol Omics 2018, 14:389–404. [DOI] [PubMed] [Google Scholar]
- 92.Wilson DJ: The harmonic mean p-value for combining dependent tests. Proc Natl Acad Sci U S A 2019, 116:1195–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Vallejos CA, Richardson S, Marioni JC: Beyond comparisons of means: understanding changes in gene expression at the single-cell level. Genome Biol 2016, 17:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Uttam S, Wong C, Amorim IS, Jafarnej SM, Tansley SM, Yang J, Prager-Khoutorsky M, Mogil JS, Gkogkas CG, Khoutorsky A: Translational profiling of dorsal root ganglia and spinal cord in a mouse model of neuropathic pain. Neurobiology of Pain 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Ji RR, Chamessian A, Zhang YQ: Pain regulation by non-neuronal cells and inflammation. Science 2016, 354:572–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kwilasz AJ, Grace PM, Serbedzija P, Maier SF, Watkins LR: The therapeutic potential of interleukin-10 in neuroimmune diseases. Neuropharmacology 2015, 96:55–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Eijkelkamp N, Steen-Louws C, Hartgring SA, Willemen HL, Prado J, Lafeber FP, Heijnen CJ, Hack CE, van Roon JA, Kavelaars A: IL4–10 Fusion Protein Is a Novel Drug to Treat Persistent Inflammatory Pain. J Neurosci 2016, 36:7353–7363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Leung A, Gregory NS, Allen LA, Sluka KA: Regular physical activity prevents chronic pain by altering resident muscle macrophage phenotype and increasing interleukin-10 in mice. Pain 2016, 157:70–79. [DOI] [PMC free article] [PubMed] [Google Scholar]; While much work has focused on inflammatory roles of microglia and macrophages that appear to play more prominent roles in males, this work highlights the pain resolving power of macrophages, an effect that occurs in males and females.
- 99.Grace PM, Fabisiak TJ, Green-Fulgham SM, Anderson ND, Strand KA, Kwilasz AJ, Galer EL, Walker FR, Greenwood BN, Maier SF, et al. : Prior voluntary wheel running attenuates neuropathic pain. Pain 2016, 157:2012–2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mogil JS: Perspective: Equality need not be painful. Nature 2016, 535:S7. [DOI] [PubMed] [Google Scholar]