1. Introduction.
Therapeutic challenges.
The International Association for the Study of Pain (IASP) Task Force recently proposed a new definition of pain as an aversive sensory and emotional experience typically caused by, or resembling that caused by, actual or potential tissue injury [188]. Importantly, acute pain serves a critical Darwinian protective function: to initiate an escape response from noxious stimuli that in the future should be avoided for personal safety. However, chronic intractable pain is maladaptive and constitutes a widespread public health issue, significantly impairing quality of life and costing nearly $600 billion per year in the US alone [1]. Current efforts to develop novel pain therapeutics are guided by the following observations: 1) pain may arise from multiple mechanisms, and this complexity reflects the difficulty in achieving significant relief; 2) chronic pain states may reflect an important sex covariate in the development of the pain phenotype; and 3) there is a growing appreciation that secondary to tissue and nerve injury, elements of the immune system are recruited in a sex-dependent manner to influence the chronic pain phenotype. In the following sections, we will discuss aspects of these three points.
Categorization of pain phenotypes.
Mechanistically, pain states evolving into a chronic pain phenotype may be classified heuristically into four categories: (1) Nociceptive pain resulting from activation of high threshold sensory neurons (nociceptors); (2) Inflammatory pain resulting from persistent inflammation in soft tissue (viscera, fascia, muscle), joints (arthritis), or other specific tissues (e.g., dental, meningeal, bone); (3) Neuropathic pain resulting from direct (trauma, compression, ischemia) or indirect (chemotoxins, radiation, or autoimmune attacks, as with paraneoplastic syndromes) injury to the peripheral afferent nerve or ganglia; or (4) Dysfunctional/Centralized pain occurring in the absence of a noxious stimulus, detectable inflammation, or structural damage to the primary afferent [228]. It should be noted that in accord with the IASP guidelines outlined above, these four categories are associated with the generation of an aversive state accompanied by changes not only in physiology (e.g., blood pressure, hormone release [42]) but also in reward and cognition (e.g., formation of a negative association leading to avoidance, development of a positive appetitive response to drugs that diminish the negative affect [169]). Chronic pain syndromes with neuropathic etiology are often challenging to manage as they tend to be refractory to treatment with anti-inflammatory drugs, and many patients report inadequate or variable relief from commonly employed first-line therapies such as anticonvulsants and antidepressants [239]. While it is informative to consider types of pain individually from a mechanistic standpoint, many chronic pain conditions represent multiple phenotypes expressed simultaneously. For instance, effective management of cancer pain may require several functionally distinct medications to target various underlying processes. Furthermore, there is increasing support for the assertion that acute pain states secondary to tissue injury may evolve into a chronic condition with peripheral and central neuropathic components [183].
Sex as a covariate in the evolution of chronic pain.
Evidence is accumulating in support of quantitative and qualitative sex differences in pain sensitivity and analgesia. Pain syndromes with high prevalence in humans – such as arthritis, temporomandibular disorder, migraine and fibromyalgia – disproportionately affect females, occurring with significantly greater incidence than in males [158; 194]. Such disparities largely have been attributed to genetic and hormonal differences between males and females in preclinical and clinical studies, as explored in depth by several elegant reviews [20; 53; 62; 73; 74; 158; 177]. Historically, most preclinical studies of pain hypersensitivity have focused on assessment of evoked behaviors in young adult male rats or mice. This approach largely derived from perceived challenges posed by evaluating effects of estrus cycle phases (which change every 4–5 days) on nociception and analgesic responsiveness [157]. Surprisingly, evidence suggests that variability associated with different stages of the estrus cycle is no greater than that occurring intrinsically in males [15]. There is a high failure rate of analgesic investigational new drugs in clinical trials, particularly for pain conditions with greater incidence in women [17]. Thus, elucidation of the molecular underpinnings of chronic pain states in females and the mechanisms underlying sex-dependent differences in pain signaling is critical for successful development of novel therapeutics [159]. Recent efforts are emphasizing inclusion of females and of spontaneous painful disease models such as osteoarthritis in companion animals to identify novel druggable targets for pain symptoms [111; 158], although the literature still remains biased toward males [159]. Recognizing this issue, the NIH specifically mandated that studies must employ both males and females unless there are organ-specific reasons to exclude one sex or the other. Several preclinical studies suggest that interactions between the immune and nervous systems contribute to sex differences in many chronic pain syndromes, and may serve as a source of novel drug targets that are specific to either males, females or both [10; 40; 54; 133; 178; 195; 199; 206; 242]. In the present review, we provide a comprehensive synthesis of reported sex differences in neuroimmune mechanisms of pain hypersensitivity in rodent models, suggesting potential high-value targets to pursue for sex-specific treatments of chronic pain in men and women.
2. Assessment of nociception in rodent models
In humans, pain is difficult to assess reliably using objective clinical measures due to its highly subjective and individualized nature [45]. Thus, diagnosis of pain syndromes and subsequent evaluation of therapeutic efficacy relies heavily on patients’ descriptions of their pain levels, features, and location. As non-verbal organisms (infants, rodents) lack this capacity, one endpoint that can be isolated and examined easily in behavioral models is nociception, or the neural process of encoding noxious stimuli constituting the sensory, non-affective component of pain (Figure 1). For excellent reviews of pain circuitry in development and adulthood, see Treede, Fitzgerald et. al., and Basbaum and Fields [14; 77; 223]. Injury- or disease-induced pain hypersensitivity results from peripheral or central sensitization, or the increased responsiveness of nociceptive neurons in the peripheral (PNS) and central nervous systems (CNS) to normal or subthreshold primary afferent input [122], a process mediated by several mechanisms described herein. Pain hypersensitivity presents both in humans and in animals as allodynia, wherein stimuli that do not normally produce pain are perceived as painful, or hyperalgesia, a state of enhanced sensitivity to noxious stimuli that is often coupled with spontaneous (non-evoked) pain [196]. If present at the site of injury, hyperalgesia is considered as primary, while that which occurs in the surrounding area is termed secondary. The development of secondary hyperalgesia is attributed to central sensitization.
Figure 1. Nociceptive sensory primary afferent pathways.

Nociceptors specialized for detection of high-intensity mechanical, thermal and/or chemical stimuli originate in the sensory ganglia (dorsal root, trigeminal and nodose) of the peripheral nervous system generally possess small- to medium-diameter, thinly myelinated Aδ fibers or small-diameter unmyelinated C fibers and terminate predominantly in spinal superficial laminae I, II and V of the dorsal horn [180; 238]. Nociceptors can be described using the following categories: Red, Neurofilament H (NFH)+ Aβ large, high threshold mechanoreceptor (HTMR)/heat; Orange, peptidergic Aδ small/medium, HTMR/heat; Green, nonpeptidergic C small, HTMR/itch/chemical; Blue, peptidergic or nonpeptidergic C small polymodal or mechanoheat/cold; Purple, peptidergic Aδ small/medium HTMR or C small polymodal. In recent years, several subclassifications of nociceptors have been proposed, and the reader is directed to several excellent references for more detailed information on evolving designations of primary afferent sensory neuron subtypes: [48; 63; 120; 124; 125; 175; 220; 225].
Classically, nociception in animals is measured as nocifensive (reflexive withdrawal) behaviors in response to evoked stimuli. However, the low probability of clinical success for candidate analgesic molecules based on evoked endpoints alone has sparked efforts to improve the face validity of preclinical paradigms of chronic pain [235]. Several groups have pursued various methods of also capturing affective and motivational components of the pain state in rodents as well as in larger animals [27; 79; 96; 131; 146; 162; 171], although these approaches are still undergoing refinement. In contrast to a spinally-organized nociceptive reflex, the perception and expression of pain unpleasantness depend on higher order functions in cognitive and limbic regions of the brain (Figure 2). Since the affective component of a pain state manifests as spontaneous as well as time-, species- and paradigm-dependent behaviors, there is no singular assay in existence that encapsulates the human experience of pain in an animal. Nonetheless, some testing approaches can capture specific aspects of emotional and motivational responses to noxious stimuli. When utilized concurrently with evoked measures, these methods may provide a stronger assessment of candidate analgesic drug efficacy in preclinical pain models. For example, rodents in pain display coping behaviors such as licking the site of injury or emitting ultrasonic vocalizations [96]. Context-dependent approach or avoidance responses following injury include conditioned place aversion (CPA) to a location linked with an aversive experience (i.e., a pain state) and conditioned place preference (CPP) for a site in which the pain state is alleviated [170]. Similarly, operant paradigms for self-administration of analgesic drugs display motivated and goal-directed behaviors to obtain relief from pain [90; 93], although interpretation of the results is complicated if the drug itself is intrinsically rewarding. Since depression often is co-morbid with chronic pain, incorporation of assays of depression-like behaviors comprised of both evoked measures, such as the forced swim test, and spontaneous assessments of anhedonia, including motivation for naturally reinforcing substances like sucrose [221].
Figure 2. Supraspinal nociceptive circuits.

Nociceptive information transmitted via the spinal cord dorsal horn is communicated to the brain along several ascending pathways (merged together in black). Laterally projecting systems to the somatosensory (SSC) and insular cortices (IC) correspond to the classical somatosensory pathway, with a highly preserved body image mapped at several synaptic links. This tract mediates the sensory/discriminative (red) component of the pain phenotype. In contrast, medially projecting systems underlying the affective (green) and motivational (purple) aspects of pain have relatively crude somatosensory mapping and project to limbic structures appreciated for their roles in emotional responses such as the parabrachial nucleus (PBN), amygdala (AMYG), anterior cingulate cortex (ACC), nucleus accumbens (NAcc) and ventral tegmental area (VTA) [5; 33; 48; 150]. Cognitive (blue) interpretation of nociceptive information is mediated by the ACC, prefrontal cortex (PFC), and NAcc. Together, these structures contribute to pain processing by integrating information about its sensory, cognitive and affective/motivational components. The activity of ascending pathways is in turn regulated by descending facilitatory and inhibitory systems, which send projections down to the spinal cord mainly from the ACC or periaqueductal gray (PAG) by way of serotonergic neurons of the nucleus raphe magnus in the rostroventral medulla (MED) or via noradrenergic neurons in the locus coeruleus to modulate excitability of dorsal horn neurons [176].
3. Influence of sex on evoked and spontaneous nocifensive behaviors
Several rodent models of chronic pain states exhibit sex differences that parallel findings in many human disorders, with greater sensitivity to nociceptive stimuli in females (Table 1). For example, tactile allodynia is more pronounced and/or of longer duration in female rodents in nerve injury paradigms of Chronic Constriction Injury (CCI) [226; 227] (but see also [216]), partial Sciatic Nerve Ligation (pSNL) [51], Sciatic Nerve Ligation (SNL) [32; 210; 212], Intra-Articular Lysophosphatidic Acid- (IA LPA)-induced neuropathy [174] and Endoneurial injection of functionally active Myelin Basic Protein (MBP) fragment (84–104) [40]. Hyperalgesic priming, an age-dependent model of the acute to chronic pain transition characterized by the prolongation of hyperalgesia by repeated nociceptive insults, also exhibits sexual dimorphism. This paradigm elicits increased allodynia and facial grimacing in female compared with male rodents in a dural Calcitonin Gene-Related Peptide (CGRP) model of migraine [10]. Female mice also experience earlier onset of pain-related functional deficits with systemic Lipopolysaccharide (LPS) [106], IA Complete Freund’s Adjuvant (CFA)-Induced Arthritis [46], Muscle hyperalgesia [86], and Femoral Bone Cancer, correlating with faster progression of disease [64; 121]. Both mechanical and cold allodynia are more pronounced in female versus male mice in models of Multiple Sclerosis (Experimental Autoimmune Encephalitis, EAE) [185] and of Complex Regional Pain Syndrome (CRPS) [213].
Table 1.
Rodent models exhibit sex-dependent differences in neuroimmune-mediated pain hypersensitivity
| Sex | Model | Pain / Nociception | Rodent | REF |
|---|---|---|---|---|
| Females | Nerve Injury (SNI) | Tactile allodynia, withdrawal frequency | C57BL/6 mice | 24 |
| Nerve Injury (SNL) | Tactile and cold allodynia | C57BL/6, SWR/J mice; Wistar rats | 32, 210, 212 | |
| Nerve Injury (CCI) | Tactile allodynia | CD-1 mice | 226, 227 | |
| Nerve Injury (L5 radiculopathy) | Tactile allodynia | Sprague Dawley, Long Evans rats | 116 | |
| Nerve injury (SNT) | Tactile allodynia | Sprague Dawley rats | 57 | |
| Nerve injury (pSNL) | Tactile allodynia | Sprague Dawley rats | 51 | |
| Systemic bacteremia (IP LPS) | Grip force | SWR/J mice | 106 | |
| Spinal TRL4 activation (IT LPS) | Grip force | SWR/J mice | 106 | |
| Serum-transfer arthritis (K/BxN) | Grip force | C57BL/6 mice | 22 | |
| Complete Freund’s Adjuvant (CFA) | Tactile allodynia | Lewis rats | 46 | |
| Fatigue-enhanced muscle insult | Muscle hyperalgesia | C57BL/6 mice | 86 | |
| Joint neuropathy (IA LPA) | Tactile allodynia | Wistar rats | 174 | |
| Femoral Bone Cancer | Limb use | Balb/c mice | 64 | |
| Complex Regional Pain Syndrome (CRPS) | Tactile allodynia | C57BL/6 mice | 213 | |
| Migraine (IC CGRP) | Tactile allodynia, Grimace | CD-1 mice; Sprague Dawley rats; | 10 | |
| Multiple Sclerosis (EAE) | Tactile and cold allodynia | C57BL/6 mice | 185 | |
| Autoimmune Demyelination | Tactile allodynia | C57BL/6 mice | 40 | |
| Males | Complete Freund’s Adjuvant (CFA) | Tactile allodynia | Sprague Dawley rats | 26 |
| Nerve injury (SNI) | Tactile allodynia | CD-1, C57BL/6 mice | 97,205,206 | |
| Context-dependent pain hypersensitivity | Tactile allodynia | CD-1 mice | 145,207 | |
| Osteoarthritis (DMM) | Tactile allodynia | CD-1, C57BL/6 mice | 139 | |
| Spinal TLR4 activation (IT LPS) | Tactile allodynia | CD-1, C57BL/6 mice | 205,206,243 | |
| Serum-transfer arthritis (K/BxN) | Tactile allodynia | C57BL/6J mice | 242 |
In contrast, some studies report greater expression of pain hypersensitivity in males. For example, the development of allodynia following intrathecal (IT) LPS or the initiation of arthritis (K/BxN serum transfer- or intraplantar IPLT CFA-induced) is more pronounced in male mice [26; 205; 206; 242; 243], yet also see [22]. Similarly, males also exhibit increased allodynia versus females in the Spared Nerve Injury (SNI) model [97; 205; 206], yet also see [24]). The discrepancies in magnitude of allodynia reported by these studies may be explained in part by different strains of rodents or paradigms utilized. Evidence of sex differences also is emerging both in mice and in humans for the expression of cued pain-related fear memory mediated by limbic, mesolimbic, and cortical circuits [11; 153]. For example, context-dependent pain hypersensitivity is increased in males relative to females when tested by a male experimenter or when examined in an environment previously associated with an aversive tonic pain experience [145; 207].
In other injury paradigms, male and female rodents develop equivalent severity of evoked or spontaneous pain-like behaviors. Studies utilizing Collagen Antibody-Induced Arthritis (CAIA) [67], IA CFA-Induced Arthritis [66], Chemotherapy-Induced Peripheral Neuropathy (CIPN) [69], adult reincision following neonatal paw incision [165] and IPLT formalin [243] models all report allodynia of similar magnitude in males and females. Interestingly, despite sex differences observed in allodynia during arthritis or following IT LPS, both male and female mice exhibit deficits in grip strength – a widely used rheumatology measure sensitive to analgesics and a frequently reported deficit known to correlate with pain in Rheumatoid Arthritis (RA) [106; 164]. Arthritis-induced declinations of functional measures such as locomotor activity and home cage wheel running also are observed in mice of both sexes [75; 104]. Both males and females exhibit post-surgical or arthritis injury-induced grimace behaviors as well as depressed nesting and burrowing [101; 102; 209]. Likewise, sucrose consumption and social exploration following systemic delivery of low dose LPS are transiently reduced [181] or unchanged [246] in both sexes. The effects of morphine on CPP and CPA during peripheral inflammation also are not significantly different between males and females [8; 92]. In spite of similar levels of pain-like behaviors in both sexes, it is important to note that the mechanisms underlying these behaviors sometimes differ in males versus females [26; 50; 89; 130; 149; 178; 195]. Thus, caution should be exercised when drawing conclusions about positive or negative effects of treatments for pain when males and females are not stratified [158].
Influence of gonadal hormones.
Sex differences in nociceptive thresholds and opioid analgesia depend largely on organizational effects of gonadal hormone status – that is, hormone action during critical periods of gestation. Specifically, neonatal exposure to testosterone appears necessary for the phenotype of decreased nociceptive sensitivity and increased morphine analgesia observed in adult males relative to females [23; 43; 114; 117]. Nonetheless, the presence of testosterone also can exert pronociceptive actions [205]. The acute, or activational, effects of estrogens on pain and analgesia are decidedly more complex. Activational effects can vary according to the type, level, stability and route of administration of estrogens, whether they are administered alone or in combination with progestins, as well as the nociceptive paradigm utilized and the chronicity of pain state. Particular caution should be exercised in the interpretation of studies in which supraphysiological doses of these hormones are administered [52; 53].
For example, systemic administration of estradiol decreases nociceptive behaviors in the second phase (10–60 minutes post-injection) of IPLT formalin-induced acute pain in gonadectomized male or female rats [81; 115; 140] and in nerve-injured intact mice [227]. In contrast, some chronic pain states that emerge days to weeks after injury or inflammation may be exacerbated by estrogens [25; 46] or are unaffected by hormones [12]. Mu Opioid Receptor (MOR)-mediated analgesia in cycling females also depends on the phase of the estrus cycle, as morphine potency is greatest in metestrus, diestrus and proestrus phases but is lowest during estrus [107; 219]. The intricate effects of estrogens are perhaps best illustrated by the observations that estradiol suppresses the induction yet facilitates the expression of hyperalgesic priming [68; 70; 103]. By contrast, progesterone appears mainly to serve a protective function, in that it mediates pregnancy-related analgesia [192] and attenuates hyperalgesia precipitated by IPLT CFA- or Carrageenan-induced monoarthritis [189; 214], excitotoxic spinal cord injury [84] and Peripheral Diabetic Neuropathy (PDN) [126]. For in-depth discussions of these processes, the reader is directed to several extensive reviews [20; 47; 52; 105; 147; 158; 234].
Influence of stress pathways.
Similarly, stress also exerts paradoxical analgesic and hyperalgesic effects that are sexually dimorphic and likely are mediated by estradiol and testosterone [99] as well as stress hormones of the Hypothalamic Pituitary Adrenal (HPA) axis such as Corticotrophin Releasing Factor (CRF), Adrenocorticotropic Hormone (ACTH), and glucocorticoids [80; 132]. Interestingly, antisense knockdown of spinal β2 adrenergic receptors attenuates CIPN in female but not male rats, while the inverse is observed following knockdown of spinal glucocorticoid receptors [69]. Following early life stress, female rats exhibit increased central amygdala CRF-mediated visceral pain hypersensitivity [184] and augmented expression of hippocampal Tumor Necrosis Factor alpha (TNFα) and IL-6 concomitant with greater SNL-induced allodynia [32]. These findings indicate sex-specific dependence on stress mediators of the sympathetic nervous system and the HPA axis in addition to gonadal hormones.
Influence of genetics.
In a rodent model of lumbar L5 radiculopathy, female Sprague-Dawley and Long-Evans but not Holtzman rats developed more severe mechanical allodynia than their male counterparts [116]. These findings are corroborated by the observation that L5 spinal nerve transection (SNT) produced greater allodynia in female versus male Sprague-Dawley rats, but no significant sex difference in Holtzman rats [57]. Swim stress-induced analgesia (SIA) is greater in female Wistar and Spontaneously Hypertensive (SHR) but not Lewis rats [230]. In contrast, SIA is enhanced in male C57BL/6 and Swiss Webster mice compared with isogenic females [160]. Similarly, morphine antinociception also is greater in several strains of male rats and mice, as reviewed in depth [161]. While allodynia is expressed in both sexes of CD-1 mice, it is evident in male but not female C57BL/6 mice in the destabilization of the medial meniscus (DMM) model of knee osteoarthritis (OA) [139]. Of note, substrain differences of C57BL/6J versus C57BL/6N mice in nociceptive behaviors are found following IPLT formalin, but not with IPLT CFA or CCI models [28]. QTL mapping in a cross of these strains uncovered a difference between B6J versus B6N, with thermal nociception being more pronounced in males. These observations indicate that rodent strain also should be considered when drawing conclusions about sex differences in pain hypersensitivity.
4. Sex-dependent neuroimmune mechanisms of pain hypersensitivity
Tissue damage or infection initiates an immune response that can ultimately lead to a chronic pain state. Acute inflammation serves a dual purpose in that a wide variety of mediators are secreted to prevent the organism from incurring further injury and to recruit peripheral immune cells for containing and repairing the damage. Historically, these factors have been characterized as either maladaptive “pro-inflammatory” or beneficial “pro-resolving”, and are released sequentially to promote active healing. Our current understanding of pathogen- or damage associated molecular pattern-induced inflammatory responses may best be described as an organized progression of interactions between immune cells. Accordingly, the secretion of factors by each cell type influences the timing and destination of chemotaxis by another cell type [19; 29; 82]. Under typical circumstances, pain remains acute as the injury is repaired and inflammation is resolved, allowing the organism to resume homeostasis.
It is widely recognized that infiltrating as well as resident immune cells likely contribute to the transition from acute to chronic pain in instances where either the damage cannot be repaired or dysregulated inflammatory signaling continues even after the injury is resolved (see Figure 3 for definitions of immune cell types) [13]. For example, infiltrating neutrophils, macrophages and T-lymphocytes as well as activated Schwann cells and satellite cells secrete factors to communicate with resident astrocytes, microglia and oligodendrocytes in the CNS to release mediators that sensitize nociceptors. These processes in turn trigger adjacent glia and neurons to drive maintenance of hyperalgesia and allodynia [31; 98; 148; 155; 198; 200; 231; 247]. Among the molecules contributing to central sensitization are neurotransmitters (glutamate, ATP), peptide signals (cytokines, chemokines, neuropeptides), and bioactive lipids generated from cyclooxygenases (COX-1/2), 12/15-Lipoxygenases (12/15-LOX), and endocannabinoid system enzymes [87; 88; 108; 138; 236; 237; 241].
Figure 3. Immune and glial cells linked to sex differences in pain hypersensitivity.

Neuroimmune cells are generally derived from four different progenitor stem cell types (Blue circles): N, Neuroepithelial [112]; D, Mesodermal [72]; M, Myeloid [7]; L, Lymphoid [6]. Neuroepithelial cells can differentiate into Astrocytes [203; 204], Oligodendrocytes [190], or Schwann Cells [163]. Mesodermal cells can be programmed into Endothelial Cells [144]. Myeloid-derived cells include Microglia [128], Macrophages [123], Neutrophils [172], or Mast Cells [113] and are referred to as Splenocytes [127] (circled in purple). Lymphoid cells differentiate into B-Cells [4] and T-Cells [65], and collectively these cells are referred to as Lymphocytes (circled in green).
Emerging evidence supports a profound role for sex-specific immune responses that may underlie disparities in incidence of pain and other neurological disorders [18; 60; 62; 141; 186; 191; 208]. Due to specific challenges unique to each sex, the male and female immune systems have different requirements, with the female immune system specifically requiring the flexibility to allow for pregnancy without attacking the fetus or sperm required for procreation [187]. Consequently, females have larger populations of most immune cells, higher levels of immunoglobulins, and exhibit stronger responses to infection [110; 168]. Conversely in males, the Y chromosome contains multiple genes involved in epigenetic regulation of the immune system and susceptibility to autoimmune diseases [37]. While some neuroimmune interactions underly nociceptive processing in both males and females, some pain states exhibit clear sex-specific mechanisms that likely affect their responsiveness to current analgesics and adjuvant therapeutics [9].
Male-specific nociceptive mechanisms.
Chronification of pain states in males is believed to be facilitated largely by the innate immune system through neutrophil recruitment to the injury site [197] and to the spinal vasculature [156], along with CNS infiltration of monocytes and activation of microglial-neuronal crosstalk via several mechanisms [179] (Table 2). While significant spinal microgliosis is evident in both sexes of rodents following injury [3; 38; 206; 242], allodynia in males is believed to be mediated by several mechanisms including, but not limited to: stimulation of purinergic P2X4 receptors [142; 224] likely on CX3CR1- (Fractalkine receptor)-positive microglia [224; 248], phosphorylation of P38 Mitogen-Activated Protein (MAP) Kinase [100; 137; 165; 216] and release of cytokines such as Brain-Derived Neurotrophic Factor (BDNF) either from spinal microglia [206] or Dorsal Root Ganglion (DRG) nociceptors [166], acting on Tropomysin receptor kinase B (TrkB) receptors in spinal dorsal horn neurons. In a model of pain chronification, hyperalgesic priming with IT BDNF or IPLT Interleukin 6 (IL-6) is mediated by activation of spinal Dopamine D5 receptors (DRD5) in male but not female spinal neurons [149]. Furthermore, the NOD-like Receptor 3 (NLRP3) inflammasome drives IL-1β release likely from non-neuronal cells, leading to subsequent activation of neuronal Transient Receptor Potential Ankyrin 1 (TRPA1) in males but not females in a postoperative pain model of paw incision [50]. Likewise, in CCI or SNT paradigms of neuropathic pain, the cytokine TNFα mediates allodynia via spinal TNF Receptor 1 (TNFR1) only in male mice despite similar expression of allodynia in both sexes [56; 211]. TNFα and IL-1β are unchanged supraspinally in anterior cingulate cortex (ACC) following common peroneal nerve injury, indicating potential local release and site-specific involvement of these mediators. However, these studies were performed in a mixed-sex cohort of mice [135].
Table 2.
Sex-dependent cellular and molecular neuroimmune mechanisms driving pain hypersensitivity
| Sex | Model | Cell Type(s) | Mediator | Rodent | REF |
|---|---|---|---|---|---|
| Females | Nerve injury (CCI) | Schwann cells | MHC-II activation of helper Th-cells | 129/FVB mice | 91 |
| Microglia, Astrocytes | Phospho-P38 MAPK in spinal cord | CD-1 mice | 226,227 | ||
| Nerve injury (SNI) | T-cells | CD4+ and CD8+ T-cell infiltration into lumbar spinal dorsal horn | CD-1 mice | 206 | |
| Nerve injury (SNL) | Mast cells | Increased mast cell infiltration into lumbar spinal dorsal horn | SWR/J mice | 212 | |
| Nerve Injury (pSNL) | T-cells | Not specified; T-cell infiltration into DRG | Sprague Dawley rats | 49,133 | |
| Nerve Injury (L5 radiculopathy) | Not specified | Spinal NRG1, ErbB4, and TAC1; Progesterone | Sprague Dawley rats | 118,119 | |
| Femoral bone cancer | Microglia; Not specified | TLR4, CD11b, CD14; Not specified | Balb/c mice; Sprague Dawley rats | 64,121 | |
| Migraine (IC nitroglycerin) | Mast cells, Endothelial cells | P2X3 | Sprague Dawley rats | 71 | |
| Migraine (IC CGRP with priming) | Not specified | RAMP1, CLR, RCP | ICR mice; Sprague Dawley rats | 10 | |
| Fatigue-enhanced muscle insult | Lymphocytes | Lymphocyte migration to muscle | C57BL/6 mice | 86 | |
| Autoimmune Demyelination (IA MBP) | T-cells | T-cell migration into DRG and spinal cord; PLC in females | C57BL/6 mice | 40 | |
| Males | Nerve injury (SNT) | Microglia | CD40+ spinal microglia interaction with infiltrating CD4+ T-cells | Balb/c mice | 34,36 |
| Nerve injury (SNI) | T-cells | CD2+ cell migration into ipsilateral spinal dorsal horn; Th1-mediated | Sprague Dawley rats | 49 | |
| CD4+/CD8+ cell infiltration into ipsilateral spinal cord | C57BL/6 mice | 44 | |||
| Neurons | TRPV1 | C57BL/6 mice | 44 | ||
| Microglia, Macrophages | TLR4, P2X4, BDNF, Phospho-P38 MAPK | CD-1, C57BL/6 mice; Sprague Dawley rats | 142,205,206 | ||
| Nerve injury (SNL) | Microglia, Neurons | P2X4, Phospho-P38 MAPK | Sprague Dawley, Wistar rats | 100,224 | |
| Oral cancer | Neutrophils | Ly6G+ neutrophil migration to tumor | C57BL6 mice | 197 | |
| IPLT carrageenan | Neutrophils; Not specified | S100A8+ and S100A9+ neutrophil migration to spinal vasculature; gonadal hormones | Sprague Dawley, Fischer 344 FBNF1 rats | 156,214 | |
| Spinal TLR4 activation (IT dsHMGB1) | Microglia, Macrophages | Spinal TNF, IL-1b, CCL2, CxCl, CxCl2, Gfap, CD11b, females recover faster | C57BL/6 mice | 2,3 | |
| Spinal TLR4 activation (IT LPS) | Not specified | Spinal TLR4 | CD-1, C3H/HEK, C3H/HeN, C57BL/6J, C57BL/10ScNJ, C57BL/10ScSnj mice | 205,243 | |
| Serum-transfer arthritis (K/BxN) | Glia | Spinal TLR4 and TNFa, spinal TRPV1 | C57BL/6 mice | 22,242 | |
| Chemotherapy-induced Peripheral Neuropathy | Not specified | Spinal TLR4, females recover faster | C57BL/6 mice | 240 |
Thus, it is important to consider that male-specific involvement of macrophages and neuroimmune mediators in allodynia is likely dependent on the paradigm utilized, the activation of specific circuits (spinal versus supraspinal), hormone status, or be influenced by other factors such as age [94; 95; 129; 134] and strain [152; 202]. Injury-induced activation of Toll-Like Receptor 4 (TLR4) is a prominent example of this controversy. IT delivery of LPS or endogenous ligands (e.g., High Mobility Group Box 1, HMGB1) and models of Peripheral neuropathy or RA elicit spinal TLR4-dependent allodynia that in some, but not all, cases is more pronounced in males than in females [2; 195; 205; 206; 240; 242; 243]. The observed reduction in responsivity of females to spinal TLR4 activation appears to be dependent on estrogen, as ovariectomy in conjunction with testosterone replacement restores expression of TLR4-mediated allodynia in CD-1 female mice to levels comparable to that observed in intact males [205]. Estrogen also attenuates LPS-induced inflammatory signaling and prevents expression of the proinflammatory phenotype of microglia during development [229; 232; 233]. Interestingly, the sex difference observed in spinal TLR4-mediated allodynia is absent when LPS is administered either at supraspinal (intracerebroventricular, ICV) or peripheral (IPLT) sites in uninjured CD-1 mice [205], or intramuscularly (IM) in a model of non-inflammatory acidic saline-induced muscle hyperalgesia [83]. In addition, systemic delivery of LPS produces pain hypersensitivity in both male and female Sprague-Dawley rats as neonates and as adults [21], correlating with decreased expression of Oprm1 encoding MOR in the Periaqueductal Gray (PAG) [246] and IL-1β mRNA in the spinal cord, ventrolateral PAG and hippocampus [182]. These observations are consistent with the finding that intra-PAG LPS in rats significantly decreases morphine antinociception in both sexes [61]. Similarly, in both sexes of C57BL/6 mice, IPLT formalin-induced delayed tactile hypersensitivity is prevented by global deletion of TLR4 [243], and spinal blockade of HMGB1 reverses CAIA-induced mechanical allodynia [2].
Furthermore, in some paradigms, crosstalk between macrophages and sensory neurons contributes to allodynia in both sexes (Table 3). For example, IPLT Angiotensin II activates its receptors (AT2R) in peripheral Iba1(+) leukocytes, leading to TRPA1 transactivation in nociceptors concurrent with pain hypersensitivity in males and females [199]. IT delivery of BDNF elicits allodynia in both sexes of CD-1 mice [143], while IL-6 contributes to enhanced hyperalgesia in males and females following muscle injury [83] and peripheral inflammation [83; 217]. In a model of hyperalgesic priming for migraine, intracisternal (IC) IL-6-induced dural inflammation is BDNF-dependent in both male and female Sprague-Dawley rats [30]. K/BxN arthritis elicits time-dependent increases in spinal and circulating TNFα in males and females, and IPLT delivery of TNFα produces spinal Transient Receptor Potential Vanilloid 1- (TRPV1)-dependent allodynia in both sexes [22; 66].
Table 3.
Sex-independent cellular and molecular neuroimmune mechanisms driving pain hypersensitivity
| Model | Cell Type(s) | Mediator | Rodent | REF |
|---|---|---|---|---|
| Nerve injury (SNT) | Spinal glia | Spinal soluble TNF and IL-1b | Holzman rats | 211 |
| Not specified | IL-6; allodynia F = M; autotomy behavior F > M | C57BL/6 × 129/BV/SV mice | 245 | |
| Microglia | CXCR1+ microglia | C57BL/6 mice | 179 | |
| C40+ spinal microglia interaction with infiltrating CD4+ T-cells | Balb/c mice | 34,36 | ||
| T-cells | Cd4+ T-cell infiltration into ipsilateral spinal cord | Balb/c mice | 35 | |
| Nerve injury (CCI) | Microglia | Phospho-P38 MAPK in males | CD-1 mice; Sprague Dawley rats | 216 |
| CXCR1+ microglia | CD-1 mice; Sprague Dawley rats | 216, 248 | ||
| P2X4+ microglia | Sprague Dawley rats | 142 | ||
| TNFR1-induced NMDA1 activation in spinal cord, cortex | C57BL/6 mice | 56 | ||
| Macrophages | CD68, Cd11b+ macrophages; tactile allodynia F = M | C57BL/6 mice | 44 | |
| T-cells | TNFR2 essential for recovery | C57BL/6 mice | 76 | |
| Nerve injury (SNI) | Microglia | P2X7 | Sprague Dawley rats | 54 |
| Nerve injury (pSNL) | Not specified | P2X7 | C57BL/6 mice | 41 |
| Postoperative pain | DRG nneurons | PRLR in females | C57BL/6.129 mice; Sprague Dawley rats | 178 |
| Microglia | Phospho-P38 MAPK in males | Sprague Dawley rats | 165 | |
| Macrophage, Mast cells, Neutrophils, | NLRP3-dependent IL-1b release in DRG and peri-incisional skin in males; NLRP3-independent in females | C57BL/6j, C57BL/6N/129 mice | 50 | |
| Mast cells | Mrgprb2 | C57BL/6 mice | 85 | |
| Complex regional pain syndrome (CRPS) | Microglia (males), T-cells (females), B-cells (both) | Spinal IL-6, NK1 in males; delayed adaptive immune response in females | C57BL/6J mice | 89 |
| IPLT Zymosan | Not specified | TLR2 | CD-1 mice | 205 |
| IPLT CFA | Neurons | High-affinity tonic GABAA current in PAG | Sprague Dawley rats | 222 |
| Not specified | Spinal TLR4, serum, TNFa, IL-1b, and IL-6 in males | CD-1 mice; Sprague Dawley rats | 26,205 | |
| IPLT Formalin | Glia, Neurons (males); T-cells (females) | DRD1, DRD3 in females; DRD4 in males; BDNF, TLR4 in both sexes | CD-1, C57BL/6J, C57BL/6/129.P2 mice | 130,149,193 202,243 |
| Muscle Hyperalgesia | Macrophages | P2X4+ resident macrophages, TLR4, IL-6 | C57BL/6 mice | 83 |
| IPLT TNFa | Not specified | Peripheral TRPA1, central TRPV1; mixed males and females | C57BL/6J.129.SvJ, CD-1 mice | 66 |
| IPLT Angiogensin II | Macrophages, Neurons | AT2R in skin macrophages transactivates TRPA1 in neurons | C57BL/6J, FVB/NJ mice | 199 |
| Hyperalgesic Priming | Neurons | Spinal DRD5 in males; DRG PRLR in females | C57BL/6J.129, C57BL/6J mice | 149,178 |
| IT BDNF | Microglia | Spinal KCC2 downregulation | CD-1 mice | 143 |
| IT NRG-1 | Astrocytes, Neurons | Spinal ErbB | Sprague Dawley rats | 118 |
| IT LPS | Not specified | Spinal TLR4 | SWR/J mice | 106 |
| ICV LPS | Not specified | TLR4 | CD-1 mice | 205 |
| Intra-PAG LPS | Microglia | TLR4 | Sprague Dawley rats | 61 |
| IPLT LPS | Not specified | TLR4 | CD-1 mice | 205 |
| Systemic Bacteremia (IP LPS, neonatal) | Microglia, Not specified | Spinal TLR4, Spinal COX2; Downregulation of Oprm1 in PAG, PFC | Sprague Dawley rats | 21,95,246 |
| Migraine (Intracisternal IL-6) | Not specified | BDNF | Sprague Dawley rats | 30 |
| IA Carrageenan | Immune cells (in joint) | P2X2 and P2X3 in both sexes; P2X7 in females | Wistar rats | 217,218 |
| IA CFA | Not specified | TRPA1, TRPV1, TNFa | CD-1, C57BL/6J mice | 66 |
| Collagen antibody-induced arthritis (CAIA) | Glia, Neurons (spinal); Macrophages | Macrophage HMGB1/TLR4 in males; Nociceptor TLR4 in females | Balb/c mice | 2,67,195 |
Female-specific nociceptive mechanisms.
Sustained allodynia in females is thought to derive in part from the adaptive immune system via activation and infiltration of Cluster of Differentiation 4 (CD4)+ T-lymphocytes to either central [206] or peripheral sites following nerve injury [91; 133] (Table 2). Interestingly, intra-sciatic (IS) injection of MBP(84–104) elicits T-cell migration to the DRG and spinal cord concurrent with tactile allodynia in female but not in male mice, in which T cells remain localized to the sciatic nerve [40]. Voluntary wheel running attenuates EAE-induced allodynia, correlating with reduced release of inflammatory cytokines from myelin-reactive T cells and attenuated DRG neuron excitability in female but not in male mice [154]. However, a female-specific role of the adaptive immune system remains to be clarified and may be paradigm- or strain-dependent. Several investigators have demonstrated that infiltrating CD4+ T-cells also contribute to tactile hypersensitivity following SNT or SNI in male Balb/c or C57BL/6 mice and Sprague-Dawley rats, respectively [34–36; 44; 49]. CD4+ T-cells mediate reduced formalin-mediated nociceptive sensitivity and increased morphine analgesia in male compared with female CD-1 mice [193]. In addition, T regulatory cells (Tregs) are essential for recovery from CCI-induced tactile allodynia via TNFα Receptor 2 (TNFR2) in both sexes [76].
Alternatively, other immune cells may be involved in female-specific mechanisms of neuropathic pain, as the number of mast cells is increased in lumbar spinal dura mater during Intradermal (ID) Capsaicin- or IPLT Carrageenan-induced inflammation [244] as well as on the side of the thalamus receiving nociceptive input following SNL [212], concurrent with allodynia in female but not in male rodents. Mast cells also mediate ID nitroglycerin-induced hyperalgesia, which is more pronounced in female rats [71]. However, paw incision- or CFA-induced activation of the mast cell receptor Mas-Related G Protein-Coupled Receptor b2 (Mrgprb2) elicits inflammation and pain hypersensitivity that is not different between male and female mice [85], so a female-specific involvement of mast cells may depend on the pain model utilized.
Microglia are believed to drive allodynia predominantly in males, yet several reports suggest that females can switch to a microglia-dependent pathway in some models of pain hypersensitivity when adaptive immune mechanisms are suppressed [89; 205; 206]. For example, microglial P2X7 is activated in females during IA Carrageenan- or Collagen- Induced Arthritis (CIA) [173; 218] yet not following IPLT CFA monoarthritis or pSNL- or SNI-induced nerve injury [41; 54]. Female-specific progesterone-dependent upregulation of Neuregulin-1 (NRG-1) in astrocytes has been observed in a model of experimental L5 lumbar radiculopathy, while exogenous spinal delivery of NRG-1 produces allodynia in both sexes [118; 119]. Mice heterozygous for NRG-1 express sex-specific reductions in serum cytokines in conjunction with increased hotplate latency, including IL-6, IL-8 and IL-10 in females and IL-1β in males [58]. However, IL-6 may also exert a female-specific protective effect, as female IL-6 deficient mice experience increased autotomy behavior following nerve injury [245]. Other mechanisms of nociception in females include inflammation-induced activation of CNS DRD3 [130] or DRD1 receptors [149], Gamma Aminobutyric Acid Receptor subtype A (GABAA) in the PAG [222] or spinal cord [78], and Prolactin receptors (PRLR) in sensory neurons [39; 178].
5. Future directions
Chronic pain affects up to 33% of the population and surpasses cancer, diabetes and heart disease in terms of societal burden [59]. Management of persistent pain is largely an exercise of trial and error, and the scarcity of viable treatment options places undue burden on the patient [183]. There is a considerable body of evidence demonstrating that an interaction between the nervous and immune systems underlies many pain syndromes at the molecular and cellular level. Since immune cells play a major role in the development of mood disorders [151], it is likely that they also may contribute to the averse emotional experience of pain. Most clinically relevant pain states have a tonic component that is not captured by standard evoked paradigms, so continued incorporation of spontaneous and functional measures of pain behaviors in preclinical studies will be critical for a deeper understanding of sex-related differences in chronic pain states and the future development of analgesics [109; 169; 171; 215]. Taken together, studies suggest that neuroimmune signaling events altered by injury, disease, or aberrant central nociceptive processing may serve as a rich resource of novel druggable targets and of predictive biomarkers suitable for patient stratification in trials [55] examining efficacy of potential pain therapeutics.
Given the challenges of developing safe and effective drugs reaching the CNS [201], one potential therapeutic avenue is to neutralize release of cytokines and chemokines released from circulating leukocytes and/or to intercept these cells before they cross the blood-brain barrier (BBB). Accordingly, the development of monoclonal antibody-based interventions targeting immune system mediators for the treatment of cancer and autoimmune diseases has grown exponentially over the past decade [136]. As our understanding of the role of specific immune cells in the development and maintenance of central sensitization continues to evolve, some of the newer-generation FDA-approved biologics may be repurposed for treating chronic pain either of peripheral origin via systemic delivery, or by routes of administration that bypass the BBB (e.g. intranasal or intrathecal). Alternatively, humanized single-domain antibodies could be harnessed as therapeutics, diagnostic agents or as delivery devices of small molecules targeting specific leukocytes owing to their stability, small size and low production cost [16]. Nonetheless, small molecules still lead the field in percent of FDA approvals in spite of challenges encountered with safety and tolerability profiles [167]. Ultimately, it is imperative for more preclinical and clinical pain studies to draw direct comparisons between males and females. Including sex as a biological variable will allow experts both to better predict which therapeutic strategies may be effective in each sex and to achieve true progress in the discovery of novel non-opioid analgesics.
Supplementary Material
Acknowledgments.
This work was supported by NIH AR075241 (AMG); Cayman Biomedical Research Institute (CABRI) Undergraduate Research Award (ISL); NIH NS099338 and DA015353 (TLY); NIH DA035865 (MWB).
Abbreviations
- AT2R
Angiotensin 2 Receptor
- BDNF
Brain-Derived Neurotrophic Factor
- CGRP
Calcitonin Gene-Related Peptide
- S100A8
Calgranulin
- S100A9
Calgranulin B
- CIPN
Chemotherapy-induced Peripheral Neuropathy
- CCI
Chronic Constriction Injury
- Cd11b
Cluster of Differentiation 11b
- CD14
Cluster of Differentiation 14
- CD2
Cluster of Differentiation 2
- CD4
Cluster of Differentiation 4
- CD40
Cluster of Differentiation 40
- CD68
Cluster of Differentiation 68
- CD8
Cluster of Differentiation 8
- CAIA
Collagen Antibody-Induced Arthritis
- CFA
Complete Freund’s Adjuvant
- CRPS
Complex Regional Pain Syndrome
- COX2
Cyclooxygenase 2
- DMM
Destabilization of Medial Meniscus
- dsHMGB1
disulfide High Mobility Group Box 1
- DRD1
Dopamine Receptor D1
- DRD3
Dopamine Receptor D3
- DRD5
Dopamine Receptor D5
- DRG
Dorsal Root Ganglion
- ErbB
Epidermal Growth Factor receptor tyrosine-protein kinase erbB-2
- ErbB4
Erb-B2 Receptor Tyrosine Kinase 4
- EAE
Experimental Autoimmune Encephalomyelitis
- CX3CR1
Fractalkine Receptor
- GABAA
Gamma Amino Butyric Acid receptor subtype A
- Oprm1
Mu Opioid Receptor gene
- GFAP
Glial Fibrillary Acidic Protein
- CXCL1 = Groα
Growth Related Oncogene alpha
- HMGB1
High Mobility Group Box 1
- IL-1β
Interleukin 1 beta
- IL-6
Interleukin 6
- IA
Intra-Articular
- IC
Intracisternal
- ICV
Intracerebroventricular
- ID
Intradermal
- IM
Intramuscular
- IP
Intraperitoneal
- IPLT
Intraplantar
- IT
Intrathecal
- LPS
Lipopolysaccharide
- Ly6G
Lymphocyte antigen 6 complex locus G6D
- LPA
Lysophosphatidic Acid
- CXCL2
MIP2α: Macrophage Inflammatory Protein 2 alpha
- MAPK
Mitogen-Activated Protein Kinase
- Mrgbr2
Mas-Related G protein-Coupled Receptor B2
- MCP1 = CCL2
Monocyte Chemoattractant Protein 1
- MBP
Myelin Basic Protein
- NRG-1
Neuregulin-1
- NK1
Neurokinin receptor 1
- NMDAR1
N-methyl D-Aspartate Receptor 1
- NLRP3
Nod-Like Receptor Protein 3
- P2X2
P2X purinoceptor 2
- P2X3
P2X purinoceptor 3
- P2X4
P2X purinoceptor 4
- P2X7
P2X purinoceptor 7
- pSNL
partial Sciatic Nerve Ligation
- PAG
Periaqueductal Gray
- KCC2
Potassium-chloride transporter member 5
- PFC
Prefrontal Cortex
- PRLR
Prolactin Receptor
- RAMP1
Receptor Activity Modification Protein 1
- SNP
Sodium Nitroprusside
- SNI
Spared Nerve Injury
- SNL
Spinal Nerve Ligation
- SNT
Spinal Nerve Transection
- TAC1
Tachykinin Precursor 1
- TLR4
Toll-Like Receptor 4
- TRPA1
Transient Receptor Potential Ankyrin 1
- TRPV1
Transient Receptor Potential Vanilloid 1
- TNFα
Tumor Necrosis Factor alpha
- TNFR1
Tumor Necrosis Factor Receptor 1
- TNFR2
Tumor Necrosis Factor Receptor 2
Footnotes
COI statement: The authors have no competing financial interests to declare.
References.
- [1].Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research. Military medicine 2016;181(5):397–399. [DOI] [PubMed] [Google Scholar]
- [2].Agalave NM, Larsson M, Abdelmoaty S, Su J, Baharpoor A, Lundback P, Palmblad K, Andersson U, Harris H, Svensson CI. Spinal HMGB1 induces TLR4-mediated long-lasting hypersensitivity and glial activation and regulates pain-like behavior in experimental arthritis. Pain 2014;155(9):1802–1813. [DOI] [PubMed] [Google Scholar]
- [3].Agalave NM, Rudjito R, Farinotti AB, Khoonsari PE, Sandor K, Nomura Y, Szabo-Pardi TA, Urbina CM, Palada V, Price TJ, Harris HE, Burton MD, Kultima K, Svensson CI. Sex-dependent role of microglia in disulfide HMGB1-mediated mechanical hypersensitivity. Pain 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Akkaya M, Kwak K, Pierce SK. B cell memory: building two walls of protection against pathogens. Nature reviews Immunology 2020;20(4):229–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Almeida TF, Roizenblatt S, Tufik S. Afferent pain pathways: a neuroanatomical review. Brain research 2004;1000(1–2):40–56. [DOI] [PubMed] [Google Scholar]
- [6].Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nature reviews Immunology 2006;6(3):205–217. [DOI] [PubMed] [Google Scholar]
- [7].Alvarez-Errico D, Vento-Tormo R, Sieweke M, Ballestar E. Epigenetic control of myeloid cell differentiation, identity and function. Nature reviews Immunology 2015;15(1):7–17. [DOI] [PubMed] [Google Scholar]
- [8].Armendariz A, Nazarian A. Morphine antinociception on thermal sensitivity and place conditioning in male and female rats treated with intraplantar complete freund’s adjuvant. Behavioural brain research 2018;343:21–27. [DOI] [PubMed] [Google Scholar]
- [9].Averitt DL, Eidson LN, Doyle HH, Murphy AZ. Neuronal and glial factors contributing to sex differences in opioid modulation of pain. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2019;44(1):155–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Avona A, Burgos-Vega C, Burton MD, Akopian AN, Price TJ, Dussor G. Dural Calcitonin Gene-Related Peptide Produces Female-Specific Responses in Rodent Migraine Models. The Journal of neuroscience : the official journal of the Society for Neuroscience 2019;39(22):4323–4331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Baliki MN, Apkarian AV. Nociception, Pain, Negative Moods, and Behavior Selection. Neuron 2015;87(3):474–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Banik RK, Woo YC, Park SS, Brennan TJ. Strain and sex influence on pain sensitivity after plantar incision in the mouse. Anesthesiology 2006;105(6):1246–1253. [DOI] [PubMed] [Google Scholar]
- [13].Baral P, Udit S, Chiu IM. Pain and immunity: implications for host defence. Nature reviews Immunology 2019;19(7):433–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Basbaum AI, Fields HL. Endogenous pain control mechanisms: review and hypothesis. Annals of neurology 1978;4(5):451–462. [DOI] [PubMed] [Google Scholar]
- [15].Beery AK. Inclusion of females does not increase variability in rodent research studies. Curr Opin Behav Sci 2018;23:143–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Belanger K, Iqbal U, Tanha J, MacKenzie R, Moreno M, Stanimirovic D. Single-Domain Antibodies as Therapeutic and Imaging Agents for the Treatment of CNS Diseases. Antibodies (Basel) 2019;8(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Berge OG. Predictive validity of behavioural animal models for chronic pain. British journal of pharmacology 2011;164(4):1195–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Berta T, Qadri YJ, Chen G, Ji RR. Microglial Signaling in Chronic Pain with a Special Focus on Caspase 6, p38 MAP Kinase, and Sex Dependence. Journal of dental research 2016;95(10):1124–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Blaho VA, Buczynski MW, Brown CR, Dennis EA. Lipidomic analysis of dynamic eicosanoid responses during the induction and resolution of Lyme arthritis. The Journal of biological chemistry 2009;284(32):21599–21612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Bodnar RJ, Kest B. Sex differences in opioid analgesia, hyperalgesia, tolerance and withdrawal: central mechanisms of action and roles of gonadal hormones. Horm Behav 2010;58(1):72–81. [DOI] [PubMed] [Google Scholar]
- [21].Boisse L, Spencer SJ, Mouihate A, Vergnolle N, Pittman QJ. Neonatal immune challenge alters nociception in the adult rat. Pain 2005;119(1–3):133–141. [DOI] [PubMed] [Google Scholar]
- [22].Borbely E, Botz B, Bolcskei K, Kenyer T, Kereskai L, Kiss T, Szolcsanyi J, Pinter E, Csepregi JZ, Mocsai A, Helyes Z. Capsaicin-sensitive sensory nerves exert complex regulatory functions in the serum-transfer mouse model of autoimmune arthritis. Brain, behavior, and immunity 2015;45:50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Borzan J, Fuchs PN. Organizational and activational effects of testosterone on carrageenan-induced inflammatory pain and morphine analgesia. Neuroscience 2006;143(3):885–893. [DOI] [PubMed] [Google Scholar]
- [24].Bourquin AF, Suveges M, Pertin M, Gilliard N, Sardy S, Davison AC, Spahn DR, Decosterd I. Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse. Pain 2006;122(1–2):14 e11–14. [DOI] [PubMed] [Google Scholar]
- [25].Bradshaw H, Miller J, Ling Q, Malsnee K, Ruda MA. Sex differences and phases of the estrous cycle alter the response of spinal cord dynorphin neurons to peripheral inflammation and hyperalgesia. Pain 2000;85(1–2):93–99. [DOI] [PubMed] [Google Scholar]
- [26].Britch SC, Goodman AG, Wiley JL, Pondelick AM, Craft RM. Antinociceptive and Immune Effects of Delta-9-Tetrahydrocannabinol or Cannabidiol in Male Versus Female Rats with Persistent Inflammatory Pain. The Journal of pharmacology and experimental therapeutics 2020;373(3):416–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Browne LE, Latremoliere A, Lehnert BP, Grantham A, Ward C, Alexandre C, Costigan M, Michoud F, Roberson DP, Ginty DD, Woolf CJ. Time-Resolved Fast Mammalian Behavior Reveals the Complexity of Protective Pain Responses. Cell reports 2017;20(1):89–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Bryant CD, Bagdas D, Goldberg LR, Khalefa T, Reed ER, Kirkpatrick SL, Kelliher JC, Chen MM, Johnson WE, Mulligan MK, Imad Damaj M. C57BL/6 substrain differences in inflammatory and neuropathic nociception and genetic mapping of a major quantitative trait locus underlying acute thermal nociception. Molecular pain 2019;15:1744806918825046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Buckley CD, Gilroy DW, Serhan CN, Stockinger B, Tak PP. The resolution of inflammation. Nature reviews Immunology 2013;13(1):59–66. [DOI] [PubMed] [Google Scholar]
- [30].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(12):2722–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Burke NN, Fan CY, Trang T. Microglia in health and pain: impact of noxious early life events. Exp Physiol 2016;101(8):1003–1021. [DOI] [PubMed] [Google Scholar]
- [32].Burke NN, Llorente R, Marco EM, Tong K, Finn DP, Viveros MP, Roche M. Maternal deprivation is associated with sex-dependent alterations in nociceptive behavior and neuroinflammatory mediators in the rat following peripheral nerve injury. The journal of pain : official journal of the American Pain Society 2013;14(10):1173–1184. [DOI] [PubMed] [Google Scholar]
- [33].Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nat Rev Neurosci 2013;14(7):502–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Cao L, Beaulac H, Eurich A. Differential lumbar spinal cord responses among wild type, CD4 knockout, and CD40 knockout mice in spinal nerve L5 transection-induced neuropathic pain. Molecular pain 2012;8:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Cao L, DeLeo JA. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. European journal of immunology 2008;38(2):448–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Cao L, Palmer CD, Malon JT, De Leo JA. Critical role of microglial CD40 in the maintenance of mechanical hypersensitivity in a murine model of neuropathic pain. European journal of immunology 2009;39(12):3562–3569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Case LK, Wall EH, Dragon JA, Saligrama N, Krementsov DN, Moussawi M, Zachary JF, Huber SA, Blankenhorn EP, Teuscher C. The Y chromosome as a regulatory element shaping immune cell transcriptomes and susceptibility to autoimmune disease. Genome Res 2013;23(9):1474–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Chen G, Luo X, Qadri MY, Berta T, Ji RR. Sex-Dependent Glial Signaling in Pathological Pain: Distinct Roles of Spinal Microglia and Astrocytes. Neuroscience bulletin 2018;34(1):98–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Chen Y, Moutal A, Navratilova E, Kopruszinski C, Yue X, Ikegami M, Chow M, Kanazawa I, Bellampalli SS, Xie J, Patwardhan A, Rice K, Fields H, Akopian A, Neugebauer V, Dodick D, Khanna R, Porreca F. The prolactin receptor long isoform regulates nociceptor sensitization and opioid-induced hyperalgesia selectively in females. Science translational medicine 2020;12(529). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Chernov AV, Hullugundi SK, Eddinger KA, Dolkas J, Remacle AG, Angert M, James BP, Yaksh TL, Strongin AY, Shubayev VI. A myelin basic protein fragment induces sexually dimorphic transcriptome signatures of neuropathic pain in mice. The Journal of biological chemistry 2020;295(31):10807–10821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, Egerton J, Murfin M, Richardson J, Peck WL, Grahames CB, Casula MA, Yiangou Y, Birch R, Anand P, Buell GN. Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain 2005;114(3):386–396. [DOI] [PubMed] [Google Scholar]
- [42].Chiang MC, Bowen A, Schier LA, Tupone D, Uddin O, Heinricher MM. Parabrachial Complex: A Hub for Pain and Aversion. The Journal of neuroscience : the official journal of the Society for Neuroscience 2019;39(42):8225–8230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Cicero TJ, Nock B, O’Connor L, Meyer ER. Role of steroids in sex differences in morphine-induced analgesia: activational and organizational effects. The Journal of pharmacology and experimental therapeutics 2002;300(2):695–701. [DOI] [PubMed] [Google Scholar]
- [44].Cobos EJ, Nickerson CA, Gao F, Chandran V, Bravo-Caparros I, Gonzalez-Cano R, Riva P, Andrews NA, Latremoliere A, Seehus CR, Perazzoli G, Nieto FR, Joller N, Painter MW, Ma CHE, Omura T, Chesler EJ, Geschwind DH, Coppola G, Rangachari M, Woolf CJ, Costigan M. Mechanistic Differences in Neuropathic Pain Modalities Revealed by Correlating Behavior with Global Expression Profiling. Cell reports 2018;22(5):1301–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Coghill RC, McHaffie JG, Yen YF. Neural correlates of interindividual differences in the subjective experience of pain. Proceedings of the National Academy of Sciences of the United States of America 2003;100(14):8538–8542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Cook CD, Nickerson MD. Nociceptive sensitivity and opioid antinociception and antihyperalgesia in Freund’s adjuvant-induced arthritic male and female rats. The Journal of pharmacology and experimental therapeutics 2005;313(1):449–459. [DOI] [PubMed] [Google Scholar]
- [47].Coraggio V, Guida F, Boccella S, Scafuro M, Paino S, Romano D, Maione S, Luongo L. Neuroimmune-Driven Neuropathic Pain Establishment: A Focus on Gender Differences. International journal of molecular sciences 2018;19(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Corder G, Castro DC, Bruchas MR, Scherrer G. Endogenous and Exogenous Opioids in Pain. Annu Rev Neurosci 2018;41:453–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, Barrett L, Brenner GJ, Vardeh D, Woolf CJ, Fitzgerald M. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. The Journal of neuroscience : the official journal of the Society for Neuroscience 2009;29(46):14415–14422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Cowie AM, Menzel AD, O’Hara C, Lawlor MW, Stucky CL. NOD-like receptor protein 3 inflammasome drives postoperative mechanical pain in a sex-dependent manner. Pain 2019;160(8):1794–1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Coyle DE, Sehlhorst CS, Mascari C. Female rats are more susceptible to the development of neuropathic pain using the partial sciatic nerve ligation (PSNL) model. Neuroscience letters 1995;186(2–3):135–138. [DOI] [PubMed] [Google Scholar]
- [52].Craft RM. Modulation of pain by estrogens. Pain 2007;132 Suppl 1:S3–12. [DOI] [PubMed] [Google Scholar]
- [53].Craft RM, Mogil JS, Aloisi AM. Sex differences in pain and analgesia: the role of gonadal hormones. European journal of pain (London, England) 2004;8(5):397–411. [DOI] [PubMed] [Google Scholar]
- [54].Dalgarno R, Leduc-Pessah H, Pilapil A, Kwok CH, Trang T. Intrathecal delivery of a palmitoylated peptide targeting Y382–384 within the P2X7 receptor alleviates neuropathic pain. Molecular pain 2018;14:1744806918795793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Davis KD, Aghaeepour N, Ahn AH, Angst MS, Borsook D, Brenton A, Burczynski ME, Crean C, Edwards R, Gaudilliere B, Hergenroeder GW, Iadarola MJ, Iyengar S, Jiang Y, Kong JT, Mackey S, Saab CY, Sang CN, Scholz J, Segerdahl M, Tracey I, Veasley C, Wang J, Wager TD, Wasan AD, Pelleymounter MA. Discovery and validation of biomarkers to aid the development of safe and effective pain therapeutics: challenges and opportunities. Nat Rev Neurol 2020;16(7):381–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Del Rivero T, Fischer R, Yang F, Swanson KA, Bethea JR. Tumor necrosis factor receptor 1 inhibition is therapeutic for neuropathic pain in males but not in females. Pain 2019;160(4):922–931. [DOI] [PubMed] [Google Scholar]
- [57].DeLeo JA, Rutkowski MD. Gender differences in rat neuropathic pain sensitivity is dependent on strain. Neuroscience letters 2000;282(3):197–199. [DOI] [PubMed] [Google Scholar]
- [58].Desbonnet L, Cox R, Tighe O, Lai D, Harvey RP, Waddington JL, O’Tuathaigh CM. Altered cytokine profile, pain sensitivity, and stress responsivity in mice with co-disruption of the developmental genes Neuregulin-1xDISC1. Behavioural brain research 2017;320:113–118. [DOI] [PubMed] [Google Scholar]
- [59].Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet (London, England) 2018;392(10159):1789–1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Donvito G, Nass SR, Wilkerson JL, Curry ZA, Schurman LD, Kinsey SG, Lichtman AH. The Endogenous Cannabinoid System: A Budding Source of Targets for Treating Inflammatory and Neuropathic Pain. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2018;43(1):52–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Doyle HH, Eidson LN, Sinkiewicz DM, Murphy AZ. Sex Differences in Microglia Activity within the Periaqueductal Gray of the Rat: A Potential Mechanism Driving the Dimorphic Effects of Morphine. The Journal of neuroscience : the official journal of the Society for Neuroscience 2017;37(12):3202–3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Doyle HH, Murphy AZ. Sex differences in innate immunity and its impact on opioid pharmacology. Journal of neuroscience research 2017;95(1–2):487–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. The Journal of clinical investigation 2010;120(11):3760–3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Falk S, Uldall M, Appel C, Ding M, Heegaard AM. Influence of sex differences on the progression of cancer-induced bone pain. Anticancer research 2013;33(5):1963–1969. [PubMed] [Google Scholar]
- [65].Farber DL. Form and function for T cells in health and disease. Nature reviews Immunology 2020;20(2):83–84. [DOI] [PubMed] [Google Scholar]
- [66].Fernandes ES, Russell FA, Spina D, McDougall JJ, Graepel R, Gentry C, Staniland AA, Mountford DM, Keeble JE, Malcangio M, Bevan S, Brain SD. A distinct role for transient receptor potential ankyrin 1, in addition to transient receptor potential vanilloid 1, in tumor necrosis factor alpha-induced inflammatory hyperalgesia and Freund’s complete adjuvant-induced monarthritis. Arthritis and rheumatism 2011;63(3):819–829. [DOI] [PubMed] [Google Scholar]
- [67].Fernandez-Zafra T, Gao T, Jurczak A, Sandor K, Hore Z, Agalave NM, Su J, Estelius J, Lampa J, Hokfelt T, Wiesenfeld-Hallin Z, Xu X, Denk F, Svensson CI. Exploring the transcriptome of resident spinal microglia after collagen antibody-induced arthritis. Pain 2019;160(1):224–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Ferrari LF, Araldi D, Green P, Levine JD. Age-Dependent Sexual Dimorphism in Susceptibility to Develop Chronic Pain in the Rat. Neuroscience 2018;387:170–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Ferrari LF, Araldi D, Green PG, Levine JD. Marked sexual dimorphism in neuroendocrine mechanisms for the exacerbation of paclitaxel-induced painful peripheral neuropathy by stress. Pain 2020;161(4):865–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Ferrari LF, Araldi D, Levine JD. Regulation of Expression of Hyperalgesic Priming by Estrogen Receptor alpha in the Rat. The journal of pain : official journal of the American Pain Society 2017;18(5):574–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Ferrari LF, Levine JD, Green PG. Mechanisms mediating nitroglycerin-induced delayed-onset hyperalgesia in the rat. Neuroscience 2016;317:121–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Ferretti E, Hadjantonakis AK. Mesoderm specification and diversification: from single cells to emergent tissues. Curr Opin Cell Biol 2019;61:110–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Fillingim RB, Gear RW. Sex differences in opioid analgesia: clinical and experimental findings. European journal of pain (London, England) 2004;8(5):413–425. [DOI] [PubMed] [Google Scholar]
- [74].Fillingim RB, King CD, Ribeiro-Dasilva MC, Rahim-Williams B, Riley JL 3rd. Sex, gender, and pain: a review of recent clinical and experimental findings. The journal of pain : official journal of the American Pain Society 2009;10(5):447–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Fischer BD, Adeyemo A, O’Leary ME, Bottaro A. Animal models of rheumatoid pain: experimental systems and insights. Arthritis research & therapy 2017;19(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Fischer R, Sendetski M, Del Rivero T, Martinez GF, Bracchi-Ricard V, Swanson KA, Pruzinsky EK, Delguercio N, Rosalino MJ, Padutsch T, Kontermann RE, Pfizenmaier K, Bethea JR. TNFR2 promotes Treg-mediated recovery from neuropathic pain across sexes. Proceedings of the National Academy of Sciences of the United States of America 2019;116(34):17045–17050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Fitzgerald M The development of nociceptive circuits. Nat Rev Neurosci 2005;6(7):507–520. [DOI] [PubMed] [Google Scholar]
- [78].Franco-Enzastiga U, Garcia G, Murbartian J, Gonzalez-Barrios R, Salinas-Abarca AB, Sanchez-Hernandez B, Tavares-Ferreira D, Herrera LA, Barragan-Iglesias P, Delgado-Lezama R, Price TJ, Granados-Soto V. Sex-dependent pronociceptive role of spinal alpha5 -GABAA receptor and its epigenetic regulation in neuropathic rodents. Journal of neurochemistry 2020. [DOI] [PubMed] [Google Scholar]
- [79].Fried NT, Chamessian A, Zylka MJ, Abdus-Saboor I. Improving pain assessment in mice and rats with advanced videography and computational approaches. Pain 2020;161(7):1420–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Gamaro GD, Torres IL, Laste G, Fontella FU, Silveira PP, Manoli LP, Frantz F, Eickhoff F, Dalmaz C. Gender-dependent effect on nociceptive response induced by chronic variable stress. Physiology & behavior 2014;135:44–48. [DOI] [PubMed] [Google Scholar]
- [81].Gaumond I, Arsenault P, Marchand S. Specificity of female and male sex hormones on excitatory and inhibitory phases of formalin-induced nociceptive responses. Brain research 2005;1052(1):105–111. [DOI] [PubMed] [Google Scholar]
- [82].Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nature reviews Immunology 2020;20(2):95–112. [DOI] [PubMed] [Google Scholar]
- [83].Gong WY, Abdelhamid RE, Carvalho CS, Sluka KA. Resident Macrophages in Muscle Contribute to Development of Hyperalgesia in a Mouse Model of Noninflammatory Muscle Pain. The journal of pain : official journal of the American Pain Society 2016;17(10):1081–1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Gorman AL, Yu CG, Ruenes GR, Daniels L, Yezierski RP. Conditions affecting the onset, severity, and progression of a spontaneous pain-like behavior after excitotoxic spinal cord injury. The journal of pain : official journal of the American Pain Society 2001;2(4):229–240. [DOI] [PubMed] [Google Scholar]
- [85].Green DP, Limjunyawong N, Gour N, Pundir P, Dong X. A Mast-Cell-Specific Receptor Mediates Neurogenic Inflammation and Pain. Neuron 2019;101(3):412–420 e413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Gregory NS, Gibson-Corley K, Frey-Law L, Sluka KA. Fatigue-enhanced hyperalgesia in response to muscle insult: induction and development occur in a sex-dependent manner. Pain 2013;154(12):2668–2676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Gregus AM, Buczynski MW, Dumlao DS, Norris PC, Rai G, Simeonov A, Maloney DJ, Jadhav A, Xu Q, Wei SC, Fitzsimmons BL, Dennis EA, Yaksh TL. Inhibition of spinal 15-LOX-1 attenuates TLR4-dependent, nonsteroidal anti-inflammatory drug-unresponsive hyperalgesia in male rats. Pain 2018;159(12):2620–2629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Gregus AM, Doolen S, Dumlao DS, Buczynski MW, Takasusuki T, Fitzsimmons BL, Hua XY, Taylor BK, Dennis EA, Yaksh TL. Spinal 12-lipoxygenase-derived hepoxilin A3 contributes to inflammatory hyperalgesia via activation of TRPV1 and TRPA1 receptors. Proceedings of the National Academy of Sciences of the United States of America 2012;109(17):6721–6726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Guo TZ, Shi X, Li WW, Wei T, Clark JD, Kingery WS. Sex differences in the temporal development of pronociceptive immune responses in the tibia fracture mouse model. Pain 2019;160(9):2013–2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Gutierrez T, Crystal JD, Zvonok AM, Makriyannis A, Hohmann AG. Self-medication of a cannabinoid CB2 agonist in an animal model of neuropathic pain. Pain 2011;152(9):1976–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Hartlehnert M, Derksen A, Hagenacker T, Kindermann D, Schafers M, Pawlak M, Kieseier BC, Meyer Zu Horste G. Schwann cells promote post-traumatic nerve inflammation and neuropathic pain through MHC class II. Scientific reports 2017;7(1):12518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Harton LR, Richardson JR, Armendariz A, Nazarian A. Dissociation of morphine analgesic effects in the sensory and affective components of formalin-induced spontaneous pain in male and female rats. Brain research 2017;1658:36–41. [DOI] [PubMed] [Google Scholar]
- [93].Hipolito L, Wilson-Poe A, Campos-Jurado Y, Zhong E, Gonzalez-Romero J, Virag L, Whittington R, Comer SD, Carlton SM, Walker BM, Bruchas MR, Moron JA. Inflammatory Pain Promotes Increased Opioid Self-Administration: Role of Dysregulated Ventral Tegmental Area mu Opioid Receptors. The Journal of neuroscience : the official journal of the Society for Neuroscience 2015;35(35):12217–12231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Hodyl NA, Walker FR, Krivanek KM, Clifton VL, Hodgson DM. Prenatal endotoxin exposure alters behavioural pain responses to lipopolysaccharide in adult offspring. Physiology & behavior 2010;100(2):143–147. [DOI] [PubMed] [Google Scholar]
- [95].Hsieh CT, Lee YJ, Dai X, Ojeda NB, Lee HJ, Tien LT, Fan LW. Systemic Lipopolysaccharide-Induced Pain Sensitivity and Spinal Inflammation Were Reduced by Minocycline in Neonatal Rats. International journal of molecular sciences 2018;19(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Huang T, Lin SH, Malewicz NM, Zhang Y, Zhang Y, Goulding M, LaMotte RH, Ma Q. Identifying the pathways required for coping behaviours associated with sustained pain. Nature 2019;565(7737):86–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].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. Pharmacological research 2019;139:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Ji RR, Chamessian A, Zhang YQ. Pain regulation by non-neuronal cells and inflammation. Science 2016;354(6312):572–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Ji Y, Hu B, Li J, Traub RJ. Opposing Roles of Estradiol and Testosterone on Stress-Induced Visceral Hypersensitivity in Rats. The journal of pain : official journal of the American Pain Society 2018;19(7):764–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Jin SX, Zhuang ZY, Woolf CJ, Ji RR. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. The Journal of neuroscience : the official journal of the Society for Neuroscience 2003;23(10):4017–4022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Jirkof P, Cesarovic N, Rettich A, Nicholls F, Seifert B, Arras M. Burrowing behavior as an indicator of post-laparotomy pain in mice. Front Behav Neurosci 2010;4:165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Jirkof P, Fleischmann T, Cesarovic N, Rettich A, Vogel J, Arras M. Assessment of postsurgical distress and pain in laboratory mice by nest complexity scoring. Laboratory animals 2013;47(3):153–161. [DOI] [PubMed] [Google Scholar]
- [103].Joseph EK, Parada CA, Levine JD. Hyperalgesic priming in the rat demonstrates marked sexual dimorphism. Pain 2003;105(1–2):143–150. [DOI] [PubMed] [Google Scholar]
- [104].Kandasamy R, Calsbeek JJ, Morgan MM. Home cage wheel running is an objective and clinically relevant method to assess inflammatory pain in male and female rats. J Neurosci Methods 2016;263:115–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Kandasamy R, Price TJ. The pharmacology of nociceptor priming. Handbook of experimental pharmacology 2015;227:15–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Kehl LJ, Kovacs KJ, Larson AA. Tolerance develops to the effect of lipopolysaccharides on movement-evoked hyperalgesia when administered chronically by a systemic but not an intrathecal route. Pain 2004;111(1–2):104–115. [DOI] [PubMed] [Google Scholar]
- [107].Kepler KL, Kest B, Kiefel JM, Cooper ML, Bodnar RJ. Roles of gender, gonadectomy and estrous phase in the analgesic effects of intracerebroventricular morphine in rats. Pharmacology, biochemistry, and behavior 1989;34(1):119–127. [DOI] [PubMed] [Google Scholar]
- [108].Khoutorsky A, Price TJ. Translational Control Mechanisms in Persistent Pain. Trends Neurosci 2018;41(2):100–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].King T, Porreca F. Preclinical assessment of pain: improving models in discovery research. Curr Top Behav Neurosci 2014;20:101–120. [DOI] [PubMed] [Google Scholar]
- [110].Klein SL. Immune cells have sex and so should journal articles. Endocrinology 2012;153(6):2544–2550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Klinck MP, Mogil JS, Moreau M, Lascelles BDX, Flecknell PA, Poitte T, Troncy E. Translational pain assessment: could natural animal models be the missing link? Pain 2017;158(9):1633–1646. [DOI] [PubMed] [Google Scholar]
- [112].Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci 2009;32:149–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Krystel-Whittemore M, Dileepan KN, Wood JG. Mast Cell: A Multi-Functional Master Cell. Frontiers in immunology 2015;6:620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Krzanowska EK, Ogawa S, Pfaff DW, Bodnar RJ. Reversal of sex differences in morphine analgesia elicited from the ventrolateral periaqueductal gray in rats by neonatal hormone manipulations. Brain research 2002;929(1):1–9. [DOI] [PubMed] [Google Scholar]
- [115].Kuba T, Wu HB, Nazarian A, Festa ED, Barr GA, Jenab S, Inturrisi CE, Quinones-Jenab V. Estradiol and progesterone differentially regulate formalin-induced nociception in ovariectomized female rats. Horm Behav 2006;49(4):441–449. [DOI] [PubMed] [Google Scholar]
- [116].LaCroix-Fralish ML, Rutkowski MD, Weinstein JN, Mogil JS, Deleo JA. The magnitude of mechanical allodynia in a rodent model of lumbar radiculopathy is dependent on strain and sex. Spine 2005;30(16):1821–1827. [DOI] [PubMed] [Google Scholar]
- [117].LaCroix-Fralish ML, Tawfik VL, DeLeo JA. The organizational and activational effects of sex hormones on tactile and thermal hypersensitivity following lumbar nerve root injury in male and female rats. Pain 2005;114(1–2):71–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Lacroix-Fralish ML, Tawfik VL, Nutile-McMenemy N, Deleo JA. Neuregulin 1 is a pronociceptive cytokine that is regulated by progesterone in the spinal cord: implications for sex specific pain modulation. European journal of pain (London, England) 2008;12(1):94–103. [DOI] [PubMed] [Google Scholar]
- [119].LaCroix-Fralish ML, Tawfik VL, Spratt KF, DeLeo JA. Sex differences in lumbar spinal cord gene expression following experimental lumbar radiculopathy. Journal of molecular neuroscience : MN 2006;30(3):283–295. [DOI] [PubMed] [Google Scholar]
- [120].Lallemend F, Ernfors P. Molecular interactions underlying the specification of sensory neurons. Trends Neurosci 2012;35(6):373–381. [DOI] [PubMed] [Google Scholar]
- [121].Lan LS, Ping YJ, Na WL, Miao J, Cheng QQ, Ni MZ, Lei L, Fang LC, Guang RC, Jin Z, Wei L. Down-regulation of Toll-like receptor 4 gene expression by short interfering RNA attenuates bone cancer pain in a rat model. Molecular pain 2010;6:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. The journal of pain : official journal of the American Pain Society 2009;10(9):895–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Lavin Y, Mortha A, Rahman A, Merad M. Regulation of macrophage development and function in peripheral tissues. Nature reviews Immunology 2015;15(12):731–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Lawson SN, Fang X, Djouhri L. Nociceptor subtypes and their incidence in rat lumbar dorsal root ganglia (DRGs): focussing on C-polymodal nociceptors, Abeta-nociceptors, moderate pressure receptors and their receptive field depths. Curr Opin Physiol 2019;11:125–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Le Pichon CE, Chesler AT. The functional and anatomical dissection of somatosensory subpopulations using mouse genetics. Front Neuroanat 2014;8:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Leonelli E, Bianchi R, Cavaletti G, Caruso D, Crippa D, Garcia-Segura LM, Lauria G, Magnaghi V, Roglio I, Melcangi RC. Progesterone and its derivatives are neuroprotective agents in experimental diabetic neuropathy: a multimodal analysis. Neuroscience 2007;144(4):1293–1304. [DOI] [PubMed] [Google Scholar]
- [127].Lewis SM, Williams A, Eisenbarth SC. Structure and function of the immune system in the spleen. Sci Immunol 2019;4(33). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nature reviews Immunology 2018;18(4):225–242. [DOI] [PubMed] [Google Scholar]
- [129].Lima M, Malheiros J, Negrigo A, Tescarollo F, Medeiros M, Suchecki D, Tannus A, Guinsburg R, Covolan L. Sex-related long-term behavioral and hippocampal cellular alterations after nociceptive stimulation throughout postnatal development in rats. Neuropharmacology 2014;77:268–276. [DOI] [PubMed] [Google Scholar]
- [130].Liu P, Xing B, Chu Z, Liu F, Lei G, Zhu L, Gao Y, Chen T, Dang YH. Dopamine D3 receptor knockout mice exhibit abnormal nociception in a sex-different manner. Journal of neuroscience research 2017;95(7):1438–1445. [DOI] [PubMed] [Google Scholar]
- [131].Liu SS, Pickens S, Burma NE, Ibarra-Lecue I, Yang H, Xue L, Cook C, Hakimian JK, Severino AL, Lueptow L, Komarek K, Taylor AMW, Olmstead MC, Carroll FI, Bass CE, Andrews AM, Walwyn W, Trang T, Evans CJ, Leslie FM, Cahill CM. Kappa Opioid Receptors Drive a Tonic Aversive Component of Chronic Pain. The Journal of neuroscience : the official journal of the Society for Neuroscience 2019;39(21):4162–4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Long CC, Sadler KE, Kolber BJ. Hormonal and molecular effects of restraint stress on formalin-induced pain-like behavior in male and female mice. Physiology & behavior 2016;165:278–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].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. Scientific reports 2017;7(1):16460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Loram LC, Sholar PW, Taylor FR, Wiesler JL, Babb JA, Strand KA, Berkelhammer D, Day HE, Maier SF, Watkins LR. Sex and estradiol influence glial pro-inflammatory responses to lipopolysaccharide in rats. Psychoneuroendocrinology 2012;37(10):1688–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Lu JS, Song Q, Zhang MM, Zhuo M. No requirement of interlukine-1 for long-term potentiation in the anterior cingulate cortex of adult mice. Molecular pain 2018;14:1744806918765799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, Wu HC. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 2020;27(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Luo X, Fitzsimmons B, Mohan A, Zhang L, Terrando N, Kordasiewicz H, Ji RR. Intrathecal administration of antisense oligonucleotide against p38alpha but not p38beta MAP kinase isoform reduces neuropathic and postoperative pain and TLR4-induced pain in male mice. Brain, behavior, and immunity 2018;72:34–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Malan TP Jr., Porreca F. Lipid mediators regulating pain sensitivity. Prostaglandins Other Lipid Mediat 2005;77(1–4):123–130. [DOI] [PubMed] [Google Scholar]
- [139].Malfait AM, Ritchie J, Gil AS, Austin JS, Hartke J, Qin W, Tortorella MD, Mogil JS. ADAMTS-5 deficient mice do not develop mechanical allodynia associated with osteoarthritis following medial meniscal destabilization. Osteoarthritis and cartilage 2010;18(4):572–580. [DOI] [PubMed] [Google Scholar]
- [140].Mannino CA, South SM, Quinones-Jenab V, Inturrisi CE. Estradiol replacement in ovariectomized rats is antihyperalgesic in the formalin test. The journal of pain : official journal of the American Pain Society 2007;8(4):334–342. [DOI] [PubMed] [Google Scholar]
- [141].Mapplebeck JC, Beggs S, Salter MW. Sex differences in pain: a tale of two immune cells. Pain 2016;157 Suppl 1:S2–6. [DOI] [PubMed] [Google Scholar]
- [142].Mapplebeck JCS, Dalgarno R, Tu Y, Moriarty O, Beggs S, Kwok CHT, Halievski K, Assi S, Mogil JS, Trang T, Salter MW. Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats. Pain 2018;159(9):1752–1763. [DOI] [PubMed] [Google Scholar]
- [143].Mapplebeck JCS, Lorenzo LE, Lee KY, Gauthier C, Muley MM, De Koninck Y, Prescott SA, Salter MW. Chloride Dysregulation through Downregulation of KCC2 Mediates Neuropathic Pain in Both Sexes. Cell reports 2019;28(3):590–596 e594. [DOI] [PubMed] [Google Scholar]
- [144].Marcelo KL, Goldie LC, Hirschi KK. Regulation of endothelial cell differentiation and specification. Circulation research 2013;112(9):1272–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Martin LJ, Acland EL, Cho C, Gandhi W, Chen D, Corley E, Kadoura B, Levy T, Mirali S, Tohyama S, Khan S, MacIntyre LC, Carlson EN, Schweinhardt P, Mogil JS. Male-Specific Conditioned Pain Hypersensitivity in Mice and Humans. Curr Biol 2020;30(3):556–559. [DOI] [PubMed] [Google Scholar]
- [146].Massaly N, Copits BA, Wilson-Poe AR, Hipolito L, Markovic T, Yoon HJ, Liu S, Walicki MC, Bhatti DL, Sirohi S, Klaas A, Walker BM, Neve R, Cahill CM, Shoghi KI, Gereau RWt, McCall JG, Al-Hasani R, Bruchas MR, Moron JA. Pain-Induced Negative Affect Is Mediated via Recruitment of The Nucleus Accumbens Kappa Opioid System. Neuron 2019;102(3):564–573 e566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].McEwen BS, Kalia M. The role of corticosteroids and stress in chronic pain conditions. Metabolism: clinical and experimental 2010;59 Suppl 1:S9–15. [DOI] [PubMed] [Google Scholar]
- [148].McMahon SB, Cafferty WB, Marchand F. Immune and glial cell factors as pain mediators and modulators. Experimental neurology 2005;192(2):444–462. [DOI] [PubMed] [Google Scholar]
- [149].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. The Journal of neuroscience : the official journal of the Society for Neuroscience 2018;38(2):379–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Melzack R, Casey KL. Localized temperature changes evoked in the brain by somatic stimulation. Experimental neurology 1967;17(3):276–292. [DOI] [PubMed] [Google Scholar]
- [151].Menard C, Pfau ML, Hodes GE, Russo SJ. Immune and Neuroendocrine Mechanisms of Stress Vulnerability and Resilience. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2017;42(1):62–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Meneses G, Rosetti M, Espinosa A, Florentino A, Bautista M, Diaz G, Olvera G, Barcena B, Fleury A, Adalid-Peralta L, Lamoyi E, Fragoso G, Sciutto E. Recovery from an acute systemic and central LPS-inflammation challenge is affected by mouse sex and genetic background. PloS one 2018;13(8):e0201375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Meulders A, Vlaeyen JW. The acquisition and generalization of cued and contextual pain-related fear: an experimental study using a voluntary movement paradigm. Pain 2013;154(2):272–282. [DOI] [PubMed] [Google Scholar]
- [154].Mifflin KA, Yousuf MS, Thorburn KC, Huang J, Perez-Munoz ME, Tenorio G, Walter J, Ballanyi K, Drohomyrecky PC, Dunn SE, Kerr BJ. Voluntary wheel running reveals sex-specific nociceptive factors in murine experimental autoimmune encephalomyelitis. Pain 2019;160(4):870–881. [DOI] [PubMed] [Google Scholar]
- [155].Milligan ED, Watkins LR. Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci 2009;10(1):23–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Mitchell K, Yang HY, Tessier PA, Muhly WT, Swaim WD, Szalayova I, Keller JM, Mezey E, Iadarola MJ. Localization of S100A8 and S100A9 expressing neutrophils to spinal cord during peripheral tissue inflammation. Pain 2008;134(1–2):216–231. [DOI] [PubMed] [Google Scholar]
- [157].Mogil JS. Animal models of pain: progress and challenges. Nat Rev Neurosci 2009;10(4):283–294. [DOI] [PubMed] [Google Scholar]
- [158].Mogil JS. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci 2012;13(12):859–866. [DOI] [PubMed] [Google Scholar]
- [159].Mogil JS. Qualitative sex differences in pain processing: emerging evidence of a biased literature. Nat Rev Neurosci 2020;21(7):353–365. [DOI] [PubMed] [Google Scholar]
- [160].Mogil JS, Belknap JK. Sex and genotype determine the selective activation of neurochemically-distinct mechanisms of swim stress-induced analgesia. Pharmacology, biochemistry, and behavior 1997;56(1):61–66. [DOI] [PubMed] [Google Scholar]
- [161].Mogil JS, Chesler EJ, Wilson SG, Juraska JM, Sternberg WF. Sex differences in thermal nociception and morphine antinociception in rodents depend on genotype. Neurosci Biobehav Rev 2000;24(3):375–389. [DOI] [PubMed] [Google Scholar]
- [162].Mogil JS, Pang DSJ, Silva Dutra GG, Chambers CT. The development and use of facial grimace scales for pain measurement in animals. Neurosci Biobehav Rev 2020;116:480–493. [DOI] [PubMed] [Google Scholar]
- [163].Monk KR, Feltri ML, Taveggia C. New insights on Schwann cell development. Glia 2015;63(8):1376–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Montilla-Garcia A, Tejada MA, Perazzoli G, Entrena JM, Portillo-Salido E, Fernandez-Segura E, Canizares FJ, Cobos EJ. Grip strength in mice with joint inflammation: A rheumatology function test sensitive to pain and analgesia. Neuropharmacology 2017;125:231–242. [DOI] [PubMed] [Google Scholar]
- [165].Moriarty O, Tu Y, Sengar AS, Salter MW, Beggs S, Walker SM. 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;39(16):3081–3093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Moy JK, Szabo-Pardi T, Tillu DV, Megat S, Pradhan G, Kume M, Asiedu MN, Burton MD, Dussor G, Price TJ. Temporal and sex differences in the role of BDNF/TrkB signaling in hyperalgesic priming in mice and rats. Neurobiol Pain 2019;5:100024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Mullard A 2019 FDA drug approvals. Nat Rev Drug Discov 2020;19(2):79–84. [DOI] [PubMed] [Google Scholar]
- [168].Nalbandian G, Kovats S. Understanding sex biases in immunity: effects of estrogen on the differentiation and function of antigen-presenting cells. Immunologic research 2005;31(2):91–106. [DOI] [PubMed] [Google Scholar]
- [169].Navratilova E, Porreca F. Reward and motivation in pain and pain relief. Nature neuroscience 2014;17(10):1304–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Navratilova E, Xie JY, King T, Porreca F. Evaluation of reward from pain relief. Annals of the New York Academy of Sciences 2013;1282:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Negus SS. Core Outcome Measures in Preclinical Assessment of Candidate Analgesics. Pharmacol Rev 2019;71(2):225–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Ng LG, Ostuni R, Hidalgo A. Heterogeneity of neutrophils. Nature reviews Immunology 2019;19(4):255–265. [DOI] [PubMed] [Google Scholar]
- [173].Nieto FR, Clark AK, Grist J, Hathway GJ, Chapman V, Malcangio M. Neuron-immune mechanisms contribute to pain in early stages of arthritis. Journal of neuroinflammation 2016;13(1):96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].O’Brien MS, Philpott HTA, McDougall JJ. Targeting the Nav1.8 ion channel engenders sex-specific responses in lysophosphatidic acid-induced joint neuropathy. Pain 2019;160(1):269–278. [DOI] [PubMed] [Google Scholar]
- [175].Odem MA, Bavencoffe AG, Cassidy RM, Lopez ER, Tian J, Dessauer CW, Walters ET. Isolated nociceptors reveal multiple specializations for generating irregular ongoing activity associated with ongoing pain. Pain 2018;159(11):2347–2362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Ossipov MH, Dussor GO, Porreca F. Central modulation of pain. The Journal of clinical investigation 2010;120(11):3779–3787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Paller CJ, Campbell CM, Edwards RR, Dobs AS. Sex-based differences in pain perception and treatment. Pain medicine (Malden, Mass) 2009;10(2):289–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Patil M, Belugin S, Mecklenburg J, Wangzhou A, Paige C, Barba-Escobedo PA, Boyd JT, Goffin V, Grattan D, Boehm U, Dussor G, Price TJ, Akopian AN. Prolactin Regulates Pain Responses via a Female-Selective Nociceptor-Specific Mechanism. iScience 2019;20:449–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Peng J, Gu N, Zhou L, U BE, Murugan M, Gan WB, Wu LJ. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nature communications 2016;7:12029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Perl ER. Cutaneous polymodal receptors: characteristics and plasticity. Prog Brain Res 1996;113:21–37. [DOI] [PubMed] [Google Scholar]
- [181].Pitychoutis PM, Nakamura K, Tsonis PA, Papadopoulou-Daifoti Z. Neurochemical and behavioral alterations in an inflammatory model of depression: sex differences exposed. Neuroscience 2009;159(4):1216–1232. [DOI] [PubMed] [Google Scholar]
- [182].Posillico CK, Terasaki LS, Bilbo SD, Schwarz JM. Examination of sex and minocycline treatment on acute morphine-induced analgesia and inflammatory gene expression along the pain pathway in Sprague-Dawley rats. Biology of sex differences 2015;6:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Price TJ, Basbaum AI, Bresnahan J, Chambers JF, De Koninck Y, Edwards RR, Ji RR, Katz J, Kavelaars A, Levine JD, Porter L, Schechter N, Sluka KA, Terman GW, Wager TD, Yaksh TL, Dworkin RH. Transition to chronic pain: opportunities for novel therapeutics. Nat Rev Neurosci 2018;19(7):383–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Prusator DK, Greenwood-Van Meerveld B. Amygdala-mediated mechanisms regulate visceral hypersensitivity in adult females following early life stress: importance of the glucocorticoid receptor and corticotropin-releasing factor. Pain 2017;158(2):296–305. [DOI] [PubMed] [Google Scholar]
- [185].Rahn EJ, Iannitti T, Donahue RR, Taylor BK. Sex differences in a mouse model of multiple sclerosis: neuropathic pain behavior in females but not males and protection from neurological deficits during proestrus. Biology of sex differences 2014;5(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Rainville JR, Hodes GE. Inflaming sex differences in mood disorders. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2019;44(1):184–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Rainville JR, Tsyglakova M, Hodes GE. Deciphering sex differences in the immune system and depression. Frontiers in neuroendocrinology 2018;50:67–90. [DOI] [PubMed] [Google Scholar]
- [188].Raja SN, Carr DB, Cohen M, Finnerup NB, Flor H, Gibson S, Keefe FJ, Mogil JS, Ringkamp M, Sluka KA, Song XJ, Stevens B, Sullivan MD, Tutelman PR, Ushida T, Vader K. The revised International Association for the Study of Pain definition of pain: concepts, challenges, and compromises. Pain 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Ren K, Wei F, Dubner R, Murphy A, Hoffman GE. Progesterone attenuates persistent inflammatory hyperalgesia in female rats: involvement of spinal NMDA receptor mechanisms. Brain research 2000;865(2):272–277. [DOI] [PubMed] [Google Scholar]
- [190].Richardson WD, Kessaris N, Pringle N. Oligodendrocyte wars. Nat Rev Neurosci 2006;7(1):11–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Rosen S, Ham B, Mogil JS. Sex differences in neuroimmunity and pain. Journal of neuroscience research 2017;95(1–2):500–508. [DOI] [PubMed] [Google Scholar]
- [192].Rosen SF, Ham B, Drouin S, Boachie N, Chabot-Dore AJ, Austin JS, Diatchenko L, Mogil JS. T-Cell Mediation of Pregnancy Analgesia Affecting Chronic Pain in Mice. The Journal of neuroscience : the official journal of the Society for Neuroscience 2017;37(41):9819–9827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].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(2):358–366. [DOI] [PubMed] [Google Scholar]
- [194].Ruau D, Liu LY, Clark JD, Angst MS, Butte AJ. Sex differences in reported pain across 11,000 patients captured in electronic medical records. The journal of pain : official journal of the American Pain Society 2012;13(3):228–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Rudjito R, Agalave NM, Farinotti AB, Lundback P, Szabo-Pardi T, Price TJ, Harris HE, Burton MD, Svensson CI. Sex- and cell-dependent contribution of peripheral HMGB1 and TLR4 in arthritis-induced pain. Pain 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Sandkuhler J. Models and mechanisms of hyperalgesia and allodynia. Physiological reviews 2009;89(2):707–758. [DOI] [PubMed] [Google Scholar]
- [197].Scheff NN, Bhattacharya A, Dowse E, Dang RX, Dolan JC, Wang S, Kim H, Albertson DG, Schmidt BL. Neutrophil-Mediated Endogenous Analgesia Contributes to Sex Differences in Oral Cancer Pain. Frontiers in integrative neuroscience 2018;12:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nature neuroscience 2007;10(11):1361–1368. [DOI] [PubMed] [Google Scholar]
- [199].Shepherd AJ, Copits BA, Mickle AD, Karlsson P, Kadunganattil S, Haroutounian S, Tadinada SM, de Kloet AD, Valtcheva MV, McIlvried LA, Sheahan TD, Jain S, Ray PR, Usachev YM, Dussor G, Krause EG, Price TJ, Gereau RWt, Mohapatra DP. Angiotensin II Triggers Peripheral Macrophage-to-Sensory Neuron Redox Crosstalk to Elicit Pain. The Journal of neuroscience : the official journal of the Society for Neuroscience 2018;38(32):7032–7057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Shi Y, Shu J, Liang Z, Yuan S, Tang SJ. EXPRESS: Oligodendrocytes in HIV-associated pain pathogenesis. Molecular pain 2016;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Shukla R, Henkel ND, Alganem K, Hamoud AR, Reigle J, Alnafisah RS, Eby HM, Imami AS, Creeden JF, Miruzzi SA, Meller J, McCullumsmith RE. Signature-based approaches for informed drug repurposing: targeting CNS disorders. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Sikandar S, Minett MS, Millet Q, Santana-Varela S, Lau J, Wood JN, Zhao J. Brain-derived neurotrophic factor derived from sensory neurons plays a critical role in chronic pain. Brain : a journal of neurology 2018;141(4):1028–1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Sloan SA, Barres BA. Mechanisms of astrocyte development and their contributions to neurodevelopmental disorders. Curr Opin Neurobiol 2014;27:75–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [204].Sofroniew MV. Astrocyte Reactivity: Subtypes, States, and Functions in CNS Innate Immunity. Trends Immunol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Sorge RE, LaCroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin JS, Ritchie J, Chanda ML, Graham AC, Topham L, Beggs S, Salter MW, Mogil JS. Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. The Journal of neuroscience : the official journal of the Society for Neuroscience 2011;31(43):15450–15454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Sorge RE, Mapplebeck JC, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin JS, Sotocinal SG, Chen D, Yang M, Shi XQ, Huang H, Pillon NJ, Bilan PJ, Tu Y, Klip A, Ji RR, Zhang J, Salter MW, Mogil JS. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nature neuroscience 2015;18(8):1081–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Sorge RE, Martin LJ, Isbester KA, Sotocinal SG, Rosen S, Tuttle AH, Wieskopf JS, Acland EL, Dokova A, Kadoura B, Leger P, Mapplebeck JC, McPhail M, Delaney A, Wigerblad G, Schumann AP, Quinn T, Frasnelli J, Svensson CI, Sternberg WF, Mogil JS. Olfactory exposure to males, including men, causes stress and related analgesia in rodents. Nat Methods 2014;11(6):629–632. [DOI] [PubMed] [Google Scholar]
- [208].Sorge RE, Totsch SK. Sex Differences in Pain. Journal of neuroscience research 2017;95(6):1271–1281. [DOI] [PubMed] [Google Scholar]
- [209].Sotocinal SG, Sorge RE, Zaloum A, Tuttle AH, Martin LJ, Wieskopf JS, Mapplebeck JC, Wei P, Zhan S, Zhang S, McDougall JJ, King OD, Mogil JS. The Rat Grimace Scale: a partially automated method for quantifying pain in the laboratory rat via facial expressions. Molecular pain 2011;7:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Stokes JA, Cheung J, Eddinger K, Corr M, Yaksh TL. Toll-like receptor signaling adapter proteins govern spread of neuropathic pain and recovery following nerve injury in male mice. Journal of neuroinflammation 2013;10:148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Sweitzer S, Martin D, DeLeo JA. Intrathecal interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibits an anti-allodynic action in a rat model of neuropathic pain. Neuroscience 2001;103(2):529–539. [DOI] [PubMed] [Google Scholar]
- [212].Taiwo OB, Kovacs KJ, Sun Y, Larson AA. Unilateral spinal nerve ligation leads to an asymmetrical distribution of mast cells in the thalamus of female but not male mice. Pain 2005;114(1–2):131–140. [DOI] [PubMed] [Google Scholar]
- [213].Tajerian M, Sahbaie P, Sun Y, Leu D, Yang HY, Li W, Huang TT, Kingery W, David Clark J. Sex differences in a Murine Model of Complex Regional Pain Syndrome. Neurobiology of learning and memory 2015;123:100–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Tall JM, Crisp T. Effects of gender and gonadal hormones on nociceptive responses to intraplantar carrageenan in the rat. Neuroscience letters 2004;354(3):239–241. [DOI] [PubMed] [Google Scholar]
- [215].Tappe-Theodor A, King T, Morgan MM. Pros and Cons of Clinically Relevant Methods to Assess Pain in Rodents. Neurosci Biobehav Rev 2019;100:335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].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, behavior, and immunity 2016;55:70–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Teixeira JM, Bobinski F, Parada CA, Sluka KA, Tambeli CH. P2X3 and P2X2/3 Receptors Play a Crucial Role in Articular Hyperalgesia Development Through Inflammatory Mechanisms in the Knee Joint Experimental Synovitis. Molecular neurobiology 2017;54(8):6174–6186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Teixeira JM, Dias EV, Parada CA, Tambeli CH. Intra-Articular Blockade of P2X7 Receptor Reduces the Articular Hyperalgesia and Inflammation in the Knee Joint Synovitis Especially in Female Rats. The journal of pain : official journal of the American Pain Society 2017;18(2):132–143. [DOI] [PubMed] [Google Scholar]
- [219].Terner JM, Lomas LM, Picker MJ. Influence of estrous cycle and gonadal hormone depletion on nociception and opioid antinociception in female rats of four strains. The journal of pain : official journal of the American Pain Society 2005;6(6):372–383. [DOI] [PubMed] [Google Scholar]
- [220].Thakur M, Crow M, Richards N, Davey GI, Levine E, Kelleher JH, Agley CC, Denk F, Harridge SD, McMahon SB. Defining the nociceptor transcriptome. Front Mol Neurosci 2014;7:87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Toma W, Kyte SL, Bagdas D, Alkhlaif Y, Alsharari SD, Lichtman AH, Chen ZJ, Del Fabbro E, Bigbee JW, Gewirtz DA, Damaj MI. Effects of paclitaxel on the development of neuropathy and affective behaviors in the mouse. Neuropharmacology 2017;117:305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Tonsfeldt KJ, Suchland KL, Beeson KA, Lowe JD, Li MH, Ingram SL. Sex Differences in GABAA Signaling in the Periaqueductal Gray Induced by Persistent Inflammation. The Journal of neuroscience : the official journal of the Society for Neuroscience 2016;36(5):1669–1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [223].Treede RD. Gain control mechanisms in the nociceptive system. Pain 2016;157(6):1199–1204. [DOI] [PubMed] [Google Scholar]
- [224].Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 2003;424(6950):778–783. [DOI] [PubMed] [Google Scholar]
- [225].Usoskin D, Furlan A, Islam S, Abdo H, Lonnerberg P, Lou D, Hjerling-Leffler J, Haeggstrom J, Kharchenko O, Kharchenko PV, Linnarsson S, Ernfors P. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nature neuroscience 2015;18(1):145–153. [DOI] [PubMed] [Google Scholar]
- [226].Vacca V, Marinelli S, Pieroni L, Urbani A, Luvisetto S, Pavone F. Higher pain perception and lack of recovery from neuropathic pain in females: a behavioural, immunohistochemical, and proteomic investigation on sex-related differences in mice. Pain 2014;155(2):388–402. [DOI] [PubMed] [Google Scholar]
- [227].Vacca V, Marinelli S, Pieroni L, Urbani A, Luvisetto S, Pavone F. 17beta-estradiol counteracts neuropathic pain: a behavioural, immunohistochemical, and proteomic investigation on sex-related differences in mice. Scientific reports 2016;6:18980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Vardeh D, Mannion RJ, Woolf CJ. Toward a Mechanism-Based Approach to Pain Diagnosis. The journal of pain : official journal of the American Pain Society 2016;17(9 Suppl):T50–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Vegeto E, Bonincontro C, Pollio G, Sala A, Viappiani S, Nardi F, Brusadelli A, Viviani B, Ciana P, Maggi A. Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. The Journal of neuroscience : the official journal of the Society for Neuroscience 2001;21(6):1809–1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Vendruscolo LF, Pamplona FA, Takahashi RN. Strain and sex differences in the expression of nociceptive behavior and stress-induced analgesia in rats. Brain research 2004;1030(2):277–283. [DOI] [PubMed] [Google Scholar]
- [231].Viader A, Blankman JL, Zhong P, Liu X, Schlosburg JE, Joslyn CM, Liu QS, Tomarchio AJ, Lichtman AH, Selley DE, Sim-Selley LJ, Cravatt BF. Metabolic Interplay between Astrocytes and Neurons Regulates Endocannabinoid Action. Cell reports 2015;12(5):798–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Villa A, Gelosa P, Castiglioni L, Cimino M, Rizzi N, Pepe G, Lolli F, Marcello E, Sironi L, Vegeto E, Maggi A. Sex-Specific Features of Microglia from Adult Mice. Cell reports 2018;23(12):3501–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Villa A, Rizzi N, Vegeto E, Ciana P, Maggi A. Estrogen accelerates the resolution of inflammation in macrophagic cells. Scientific reports 2015;5:15224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [234].Vodo S, Bechi N, Petroni A, Muscoli C, Aloisi AM. Testosterone-induced effects on lipids and inflammation. Mediators of inflammation 2013;2013:183041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Whiteside GT, Pomonis JD, Kennedy JD. An industry perspective on the role and utility of animal models of pain in drug discovery. Neuroscience letters 2013;557 Pt A:65–72. [DOI] [PubMed] [Google Scholar]
- [236].Wilkerson JL, Donvito G, Grim TW, Abdullah RA, Ogasawara D, Cravatt BF, Lichtman AH. Investigation of Diacylglycerol Lipase Alpha Inhibition in the Mouse Lipopolysaccharide Inflammatory Pain Model. The Journal of pharmacology and experimental therapeutics 2017;363(3):394–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Wilkerson JL, Ghosh S, Bagdas D, Mason BL, Crowe MS, Hsu KL, Wise LE, Kinsey SG, Damaj MI, Cravatt BF, Lichtman AH. Diacylglycerol lipase beta inhibition reverses nociceptive behaviour in mouse models of inflammatory and neuropathic pain. British journal of pharmacology 2016;173(10):1678–1692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [238].Willis WD, Westlund KN. Neuroanatomy of the pain system and of the pathways that modulate pain. J Clin Neurophysiol 1997;14(1):2–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Witkin LR, Zylberger D, Mehta N, Hindenlang M, Johnson C, Kean J, Horn SD, Inturrisi CE. Patient-Reported Outcomes and Opioid Use in Outpatients With Chronic Pain. The journal of pain : official journal of the American Pain Society 2017;18(5):583–596. [DOI] [PubMed] [Google Scholar]
- [240].Woller SA, Corr M, Yaksh TL. Differences in cisplatin-induced mechanical allodynia in male and female mice. European journal of pain (London, England) 2015;19(10):1476–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].Woller SA, Eddinger KA, Corr M, Yaksh TL. An overview of pathways encoding nociception. Clinical and experimental rheumatology 2017;35 Suppl 107(5):40–46. [PMC free article] [PubMed] [Google Scholar]
- [242].Woller SA, Ocheltree C, Wong SY, Bui A, Fujita Y, Goncalves Dos Santos G, Yaksh TL, Corr M. Neuraxial TNF and IFN-beta co-modulate persistent allodynia in arthritic mice. Brain, behavior, and immunity 2019;76:151–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [243].Woller SA, Ravula SB, Tucci FC, Beaton G, Corr M, Isseroff RR, Soulika AM, Chigbrow M, Eddinger KA, Yaksh TL. Systemic TAK-242 prevents intrathecal LPS evoked hyperalgesia in male, but not female mice and prevents delayed allodynia following intraplantar formalin in both male and female mice: The role of TLR4 in the evolution of a persistent pain state. Brain, behavior, and immunity 2016;56:271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [244].Xanthos DN, Gaderer S, Drdla R, Nuro E, Abramova A, Ellmeier W, Sandkuhler J. Central nervous system mast cells in peripheral inflammatory nociception. Molecular pain 2011;7:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [245].Xu XJ, Hao JX, Andell-Jonsson S, Poli V, Bartfai T, Wiesenfeld-Hallin Z. Nociceptive responses in interleukin-6-deficient mice to peripheral inflammation and peripheral nerve section. Cytokine 1997;9(12):1028–1033. [DOI] [PubMed] [Google Scholar]
- [246].Yan S, Kentner AC. Mechanical allodynia corresponds to Oprm1 downregulation within the descending pain network of male and female rats exposed to neonatal immune challenge. Brain, behavior, and immunity 2017;63:148–159. [DOI] [PubMed] [Google Scholar]
- [247].Zarpelon AC, Rodrigues FC, Lopes AH, Souza GR, Carvalho TT, Pinto LG, Xu D, Ferreira SH, Alves-Filho JC, McInnes IB, Ryffel B, Quesniaux VF, Reverchon F, Mortaud S, Menuet A, Liew FY, Cunha FQ, Cunha TM, Verri WA Jr. Spinal cord oligodendrocyte-derived alarmin IL-33 mediates neuropathic pain. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 2016;30(1):54–65. [DOI] [PubMed] [Google Scholar]
- [248].Zhuang ZY, Kawasaki Y, Tan PH, Wen YR, Huang J, Ji RR. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain, behavior, and immunity 2007;21(5):642–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
