Skip to main content

The influence of sex on neuroimmune communication, pain, and physiology

Abstract

With the National Institutes of Health’s mandate to consider sex as a biological variable (SABV), there has been a significant increase of studies utilizing both sexes. Historically, we have known that biological sex and hormones influence immunological processes and now studies focusing on interactions between the immune, endocrine, and nervous systems are revealing sex differences that influence pain behavior and various molecular and biochemical processes. Neuroendocrine-immune interactions represent a key integrative discipline that will reveal critical processes in each field as it pertains to novel mechanisms in sex differences and necessary therapeutics. Here we appraise preclinical and clinical literature to discuss these interactions and key pathways that drive cell- and sex-specific differences in immunity, pain, and physiology.

Plain English summary

In the last decade, NIH-funded basic and clinical research studies have been mandated to include sex as a biological variable, and this has resulted in a boom of studies discovering sex differences. We know that females are more likely than males to develop chronic pain conditions and experience higher levels of pain for longer periods of time. Conditions that demonstrate this include migraine, arthritis, and peripheral neuropathy. Furthermore, these sex differences extend to surgical outcomes and are observable at the molecular, cellular, and systemic levels. Although pain perception pathways differ by sex, studies are also focusing on differences in the “conversation” between the immune system and the nervous system while addressing implications of sex hormones to gain a better understanding of the impact on pain, physiology, and behavior. Lifestyle factors such as diet and alcohol consumption also play a role in affecting the perception and impact of pain conditions. As this area of research continues to grow, the development and availability of sex-specific treatment options will grow and lead to improved patient outcomes and more effective pain management strategies.

Highlights

  • Immune cell types and molecular pathways drive sex specific differences in inflammatory and neuropathic pain.

  • TLR4 involvement in pain pathways show contextual sex-specific variations and its interaction with TRPV1 channels adds complexity to nociceptor physiology.

  • Female mice tend to exhibit stronger TRPV1-mediated pain responses compared to males due to estrogen enhancement of TRPV1 expression levels.

  • The interplay between nuclear receptors (PPARγ, PPARα) and prolactin receptors influences sex-specific neuroimmune interactions. In males, PPARα impacts immune responses and neuroinflammation. In females, PPARγ activity influences pain perception and PRL influences pain sensitivity.

  • Personalized approaches are needed for pain management in males and females due to sex differences in pain perception and treatment outcomes driven by variations in hormone levels, adiposity, and neuroimmune mechanisms of inflammation.

Neuroimmune interactions, pain, and sex differences: an overview

The study of immune-to-neural interactions, or neuroimmunology, is a burgeoning field that integrates two of the most complex organ systems of the body. With roots in immunology and neuroscience, this integration spans over 45 years of studies with influence from physiology investigators like Ivan Pavlov that hypothesized that immunity could be conditioned, and Hans Selye, who demonstrated that various stressors influence immune and endocrine effects [1,2,3,4,5]. Researchers in the 1970’s coined the term “psychoneuroimmunology” and put this research topic on the map as a pertinent integrative research necessity [6,7,8]. Over the years, the field has split into defined categories with psychology and molecular neuroscience dictating the major research focus arms [9,10,11]. A surge in interest and citations in immune-to-neural interactions happened in 2014 and coincided with the national institutes of health’s (NIH) mandate that all funded studies needed to consider sex as a biological variable (SABV) [12]. This is a logical extension because the integrative nature of neuroimmunology allows for the assessment of both behavioral and physiological output that can validate the influence of one system over the other and any apparent sex differences. This is a unique aspect of the field where the inherent experimental design allows for our understanding of these interactions and outputs at various levels (Table 1).

Table 1 Summary of the described sex differences across various immune-, neuro-, and endocrine-based targets. The denotation of “N/A” highlights interactions and mechanisms that have yet to be elucidated and posits the exploration of whether sex differences exist for these pathways

One area that has gained interest is how the nervous system and immune system communicate to modulate pain and how pain differs between sexes. Pain is a multi-faceted, subjective experience that involves the integration of peripheral sensory systems and higher brain processing centers. Within the peripheral nervous system (PNS), the dorsal root ganglia (DRG), which are a heterogeneous population of sensory neurons, innervate peripheral tissues and are the first level of integration of sensory information and mediation of responses to noxious stimuli. These nociceptive neurons, or nociceptors, are characterized by their varying degrees of myelination, size, and signaling capabilities [13]. Recent single-cell RNA-sequencing data indicates that there are at least 10 major populations of sensory neurons in the murine DRG, with numerous subtypes that further distinguish their unique physiological properties [14]. Further, spatial transcriptomics have identified at least twelve sensory neuron subtypes in human DRG [15, 16]. These neurons actively respond to different external stimuli, such as changes in thermal, mechanical, or chemical stimuli. They play a necessary role to alert the individual to potentially harmful situations or environments and initiate defensive behaviors. When activated by a noxious stimulus, DRG nociceptors transduce this signal to the dorsal horn of the spinal cord and then to the brain, including the periaqueductal gray, amygdala, hypothalamus, and other regions of the cerebral cortex, to be processed and interpreted as pain (Fig. 1).

Fig. 1
figure 1

The ascending pain pathway (Red). Noxious stimuli, such as thermal, chemical, or mechanical stimuli, or inflammatory stimuli, such as lipopolysaccharide (LPS), are detected by receptors expressed on nociceptors innervating peripheral tissue. Thermal stimuli activate TRP channels, such as TRPV1, while inflammatory stimuli activate other receptors, like activation of TLR4 by LPS. Following stimulation, nociceptors initiate pain transmission by propagating information to sensory neuron cell bodies located in dorsal root ganglia (DRG). These neurons synapse onto second order neurons within the dorsal horn of the spinal cord which in turn transmit nociceptive signals to the brain where it is interpreted as pain. Neuroimmune interactions within different levels of the ascending pain pathway influence the development and maintenance of pain in a sex- and cell-specific manner. Differences in arrow sizes indicate level of contribution, with larger arrows demonstrating greater impact, and are used to highlight sex-specific differences

Short term, or acute pain, can prompt defensive behaviors following injury to facilitate healing. Maladaptive plasticity of the nervous system can translate to various pain states [17]. Long-term, or chronic pain, is characterized by pain that persists beyond a biological benefit and is a pervasive disease state that affects over 20% of the world’s population, with an additional 10% diagnosed with chronic pain each year [18]. In the United States, the Centers for Disease Control and Prevention (CDC) lists chronic pain as the number one cause of long-term disability and loss of productivity [19]. Chronic pain has a higher prevalence in females, which account for 70% of cases; females experience increased severity and duration of pain compared to males [20]. Females are more likely to develop pain disorders that affect the musculoskeletal system such as fibromyalgia [21], osteoarthritis, low back pain [22], and migraines [23, 24]. Moreover, and to a lesser extent, females have reported more pain after invasive procedures than males [25]. Since pain is such a complex physical and emotional state, preclinical or translational studies assess the physical influence of noxious stimuli, or nociception [26].

One mechanism by which chronic pain occurs in the periphery is ectopic activity of peripheral nerves and cell bodies. During ectopic activity, pain is generated as a result of ion channel and receptor upregulation and hypersensitization to neuromodulators [27]. Specific channel subtypes, such as Transient Receptor Potential Channels (TRP) channels TRPA1 and TRPV1, are crucial components of nociceptor signaling as they regulate pain, temperature, and mechanical sensation through Ca2+ influx [28]. Activation of these TRP channels on nociceptors have been implicated in the pathogenesis of chronic pain (Fig. 1). Following injury or inflammation, TRP channels are modulated in 2 ways: (1) their expression in DRG neurons are upregulated and (2) the channels are sensitized through phosphorylation, both of which contribute to enhance neuronal activity and chronic pain [29,30,31]. This may suggest that TRP channels could be a promising candidate for pain therapeutics [32]. Dysregulation of central nociceptive circuitry can also lead to maladaptive plasticity and chronic pain. Following exposure to certain stimuli, neurons in the central nervous system (CNS) can exhibit increased responsiveness to previously subthreshold inputs from peripheral sensory neurons to enhance pain responses. While intense noxious stimuli elicit pain responses of sensory neurons, it is noteworthy that repeated or prolonged non-noxious stimuli, such as moderate alcohol consumption, short-term high-fat diet, or other dietary components such as ω-6 polyunsaturated fatty acids, also reduce pain thresholds and can contribute to the development of chronic pain [33,34,35].

The immune system plays a key role in the activation and sensitization of nociceptors. Under inflammatory conditions, such as tissue injury or infection, immune cells release signaling molecules that can stimulate nociceptors to initiate nociceptive signaling or increase expression of receptors for noxious stimuli [36]. Conversely, nociceptive neurons can also regulate immune responses and inflammation [37,38,39]. The basic mechanisms and dynamics of neuroimmune modulation of pain are under investigation; however, not all is equal between sexes considering cell activity. This review discusses neuroimmune interactions in pain physiology and how these interactions differ between sexes.

Neuroimmune interactions in pain physiology

Considerable evidence shows that immune cells play a significant active role in the onset and persistence of chronic pain conditions. Tissue resident immune cells inhabit dedicated tissue types and adapt to the metabolic function and defense of the tissue [40,41,42,43,44,45]. Tissue resident immune cells are the main actors in mediating inflammatory responses that precipitate acute and chronic pain in their efforts to preserve tissue integrity and have been demonstrated to be primary contributors to muscle pain [46]. They directly interact with and receive signals from neurons for rapid and stronger immune responses to danger signals, infections, and physiological imbalance within the tissue.

Immune cells are activated by a variety of substances such as damage-associated molecular patterns (DAMPs) released by damaged cells, cytokines, and chemical messengers released from neurons, including norepinephrine and acetylcholine (Ach), substance P and calcitonin gene related peptide (CGRP) [47,48,49,50,51]. The pro-inflammatory cytokine, Tumor Necrosis Factor-α (TNF-α) can activate and induce glutamate release from microglia in cases of oxidative stress to produce neurotoxic effects on neurons [52,53,54]. Following their activation, immune cells migrate toward damage signals from within the tissue and provide tissue protection by phagocytosis of pathogens and dead cells and by releasing factors to stimulate repair and regeneration of tissue-specific cells such as epithelial cells, fibroblasts, and neurons [55, 56]. Immune cells, such as DRG resident macrophages and natural killer (NK) cells [57,58,59,60]can also directly interact with sensory neurons to mediate pain and sensation. For example, injury to the DRG triggers a pro-inflammatory activation state of macrophages [61, 62]. Consequently, this immune response increases phagocytic activity, activates NK cells, induces cytotoxic action, and propagates pro-inflammatory cytokine release culminating in enhanced neuron responsiveness [57, 59]. This enhanced activity leads to maladaptive plasticity and sensitization of neurons to lower pain thresholds and sustain chronic pain states.

In addition to tissue-resident immune cells, circulating immune cells which travel in the blood and lymph, respond to danger signals throughout the entire body, modulate neuronal activity as part of the humoral response and vice versa through neuroimmune crosstalk [37, 63]. For example, neurons can engage circulating monocytes via expression of the chemokine C-C Motif Chemokine Ligand 2 (CCL2) which binds to C-C Motif Chemokine Receptor 2 (CCR2) on monocytes, recruiting them to sites of injury [36, 64,65,66]. Tissue infiltrated monocytes differentiate into macrophages and modulate neuronal activity directly. Circulating neutrophils and helper T-cells are also recruited to injury sites and contribute to sensory neuron signaling and sensitization by releasing pro-inflammatory cytokines, such as TNF-α, Interleukin-6 (IL-6), and Interleukin-1β (IL-1β), and other factors [67, 68]. These molecules increase neuronal excitation and alter pain sensitivity. For example, IL-1β binds to DRG sensory neurons and promotes nociceptive signaling by relieving the resting slow inactivation of sodium channels to increase sodium currents and nociceptor excitability [69, 70]. Immune cells also release anti-inflammatory cytokines such as Transforming growth factor-β1 (TGF-β1) and Interleukin-10 (IL-10), which promote sensory neuron repair and reduce neuronal signaling to abrogate pain transmission and contribute to pain resolution [71,72,73]. While these circulating immune cells can be studied via whole blood isolation and plasma analysis, tissue resident immune cells are much more difficult to study due to tissue accessibility issues and overlapping identifying markers to infiltrating circulating cells [74,75,76,77].

Other circulating immune components, such as complement proteins C3a and C5a, contribute to direct neuronal stimulation that leads to pain transduction [78,79,80]. These two complement proteins, generally referred to as anaphylatoxins, are primarily effectors of innate immunity concerned with the defense of neurons by promoting inflammation [81, 82]. C3a and C5a have a neuroprotective role against excitotoxic neuronal injury, promoting neurogenesis, inhibiting caspase-3 activity, regulation of glutamate receptor expression, and astrocyte stimulation [83,84,85,86,87]. Despite these neuroprotective effects, C3a and C5a can trigger pain responses to heat by direct modulation of neurons via activation of neuronal complement-specific receptors and TRPV1 channels [78, 88].

Important to note, this is a two-way conversation, and not only can immune cells influence neuronal activity, but neurons can regulate the activity of immune cells. Sensory neurons transmit electrochemical signals to lymph tissues and immune cells to modulate inflammatory and pain responses through molecular- and receptor-mediated communication [36, 89,90,91]. Moreover, new research has demonstrated sensory neurons participate in neuroimmune crosstalk with various tissues that were historically considered passive, including the skin, colon, lungs, and adipose tissue to drive physiology and further drive immunological responses [89, 92,93,94,95]. Immune cells express receptors for neurotransmitters that modulate their response to noxious signals and activation of these receptors on immune cells regulates the production and release of inflammatory molecules that can in turn stimulate neurons [96, 97]. For example, GABA suppresses T-cell and macrophage cytokine production capabilities, thus dampening inflammatory responses, and GABA bound to receptors on T-cells produces a profound effect by blocking intracellular calcium signaling and MAPK activation [97, 98].

Both neuronal and immune cell types undergo metabolic changes where neuronal synaptic plasticity is modified, and resident immune cells are activated and transformed morphologically [99, 100]. Further, these signals can mediate a variety of genomic and non-genomic effects that modulate pain and homeostasis [101,102,103,104,105]. Genomic effects are coordinated through intracellular receptors that function as transcription factors and influence mRNA levels for specific proteins such as cytokines, prostaglandins, and other inflammatory mediators. Membrane-bound receptors coordinate both genomic and non-genomic effects, inducing rapid signaling via ion channels, G-protein coupled receptors, and growth factor receptors that activate kinase signaling cascades [101, 106]. Both sensory neurons and immune cells express receptors for a variety of signaling molecules that can initiate these cascades, such as purinergic receptors (P2X/P2Y) [107,108,109,110], DAMPs [111], and Toll-like receptors (TLR) [111,112,113].

One example of neuroimmune communication is neuron-microglia crosstalk in the CNS. Microglia, CNS resident immune cells, survey their environments and monitor neuronal conditions. Activated microglia modulate neuronal activity to drive behaviors, like learning and memory [114,115,116,117,118], depression [119,120,121,122,123], sickness [115, 124, 125], and pain [126,127,128,129,130]. Injury can induce the release of various signaling factors from damaged cells [131,132,133]. These stimuli activate microglia and attract them to damaged areas to clear debris and dead cells. For example, glutamate levels rise under inflammatory conditions, such as a traumatic brain injury, which increases neuronal activity and contributes to the development of central sensitization and persistent pain. In addition to increasing neuronal activity, glutamate induces calcium influx which initiates ATP-ADP release from neurons leading to stimulation of microglial P2Y12 receptors [134]. Microglial P2Y12 stimulation is one of the first steps of microglial activation and induces microglia to migrate toward the site of injury. Further, ATP signaling on microglia via P2X receptors increases intracellular Ca2+ and triggers the release of cytokines and chemokines and is important for phagocytosis responses [135,136,137]. Microglia also become activated by neurotransmitters, glutamate, and acetylcholine, via NMDA/AMPA and cholinergic receptors, respectively [135, 138,139,140,141]. Glutamatergic stimulation of microglia triggers their modulation of neurons by the release of signaling factors to facilitate neuroimmune crosstalk and by directly wrapping around damaged axons [116, 139, 142,143,144,145].

The action of microglia in response to neurotransmitters can be different based on their location in the brain. For example, it has been shown that following treatment with the glutamate receptor-agonist, N-methyl-D-aspartic acid, microglia are neuroprotective in the CA3 and DG regions of the hippocampus, maintaining a ramified state in the CA3 region and “hypertrophic” morphology in the DG [146]. In contrast, in the CA1 region, microglia are not neuroprotective, adopt an amoeboid state, and NMDA- excitotoxicity induces neuronal death [146]. It is possible that these different microglial responses to NMDA may be due to different receptor subtype expression in different regions of the brain and differences in age [147].

Immune components expressed by neurons to mediate pain

Emerging research shows specific receptors and molecules participate in driving pain states differently depending on sex [101, 148]. Additionally, peripheral sensory neurons release neuropeptides, like Substance P and CGRP, that can directly activate nociceptors and enhance infiltration of circulating immune cells to act on injured nerves, causing sensitization and skewing them toward pain states [148,149,150]. Activation of Toll-Like Receptor 4 (TLR4) and NOD-Like receptor protein 3 (NLRP3) on neurons in response to pathogen-associated molecular patterns (PAMPs) and DAMPs leads to the expression of molecules that promote maladaptive plasticity of neurons and enhance their sensitization toward pain states. In injury models, sensory neuron signaling via TLR4 and NLRP3 lead to greater pain sensitivity in females compared to males, suggesting activation of these receptors produces different cellular responses [151, 152]. Pathogens release molecules, such as N-formyl peptides, that also circulate and directly activate neurons via N-formyl peptide receptors (FPR) to protect against further damage to infected areas [153] (Fig. 2).

Fig. 2
figure 2

Sexual dimorphic mechanisms in neuroimmune signaling driven by interactions with sensory neurons. In females, sensory neurons are thought to be the main drivers in pain signaling as seen by the female-skewed pathways

Immunoglobulin-like receptors (Fc) are expressed on sensory neurons and are a key component in mediating pain independent of immune cell activation or inflammation [154,155,156,157]. Fc receptors interface with circulating immunoglobulin subtypes and facilitate increases in intracellular calcium to sensitize sensory neurons or heighten nociception in females [155, 158,159,160]. In a study investigating the role of Fc receptor subtypes, FcγRII and FcγRIII, using the K/BxN model of serum-transfer arthritis, only FcγRIII-deficient mice had decreased paw inflammation [161]. Intriguingly, not only was there a lack of attenuation of paw inflammation in the FcγRII-deficient mice, but the time to onset was decreased and the severity of arthritis was increased [161]. Understanding the interactions of these immune components and their direct action on neurons to contribute to pain may highlight them as potential targets for prevention of pain.

Sex differences in neuroimmune interactions

Through pre-clinical and clinical studies, neuroimmune interactions have been shown to propagate and maintain pain states differentially in males and females [162, 163] An example of this is the NLRP3 inflammasome in inflammatory pain models, which contributes to female-specific, sensory neuron driven pain [152, 164]. In an oral cancer pain model, male-specific sexual dimorphisms were observed in neutrophil-mediated suppression of pain [165]. Microglia in the CNS are reported to possess a more male-specific role in mediating mechanisms in the development of chronic pain [166, 167], but there is new data that suggests that cell signaling has more influence than specific cell types [168].

In females, T-cells have been shown to have a greater involvement in pain development and outcomes [166, 169,170,171,172,173]. In a neuropathic injury model, T-cell infiltration into the spinal cord was shown to resolve pain in females but not males [166, 174]. Furthermore, female mice lacking T-cells did not experience pregnancy analgesia in neuropathic or inflammatory pain models [175].

A study that compared central and peripheral immune cell infiltration following nerve injury suggests sex dimorphic processes in the adaptive immune cells that infiltrate the DRG 8 days following injury, with greater B-cell infiltration in male mice and greater T-cell infiltration in female mice [176]. Peripheral nerve injury increases inflammation and pain not only at the site of injury but also in the central nervous system. One mechanism for this spread of pain signals involves colony-stimulating factor 1 (CSF1), a cytokine important for macrophage differentiation. Following peripheral neuropathic injury, CSF1 is expressed de novo in the DRG. CSF1 then travels to the spinal cord where it interacts with microglia in the dorsal horn [177]. Interestingly, research suggests that CSF1 is expressed at higher levels in females than males after neuropathic injury suggesting a sex-dependent regulation of CSF1 signaling during pain development [178]. While studies show male-specific responses to intrathecal CSF1 administration, some lack data on female responses, making direct sex comparison difficult [177, 179]. This raises the possibility that CSF1 signaling on a cellular level may be similar in both sexes, but in females, higher levels of CSF1 may contribute to differences in neuropathic pain. Additionally, if CSF1 expression levels is indeed the key to sex disparity, using the same intrathecal dose of CSF1 may mask sex differences in neuropathic pain models. Intrathecal CSF1 has shown a male-biased outcome due to microglia’s role in regulating pain states in males [127, 180, 181]. CSF1 can mediate crosstalk between lymphocytes and spinal cord microglia in females by acting as a modulator of Treg immunosuppressive activity [174]. Peripheral macrophages also express the receptor for CSF1 (CSF1R) (Fig. 3), suggesting that macrophages in addition to microglia play a role in sex-specific differences in CSF1-mediated neuropathic injury. More evidence for neuroimmune crosstalk, was described when deletion of the CSF1 ligand in sensory neurons reduced tissue resident macrophage proliferation (Ki67+) in the DRG of males but not females following neuropathic injury [182].

Fig. 3
figure 3

Sexual dimorphic mechanisms in neuroimmune signaling driven by interactions with macrophages. In males, immune cells are thought to be the main drivers in pain signaling, such as through TLR4 signaling

Additionally, other receptors such as TLR4 on different immune and neuronal populations can lead to differential pain sensitivity in males and females [183]. A study has shown that TLR4 on nociceptors drives early neuropathic pain development in a female-specific manner [151]. These data along with the NLRP3 data imply sensory neuron-derived mechanisms differentiate male and female pain development. The involvement of these receptors and immune cells to facilitate neuroimmune communication can help to determine potential mechanisms underlying pain and immunity.

TLR4: a mediator of sex-specific differences in neuroimmune interactions

Toll receptors are evolutionarily conserved proteins involved in innate immune processes that recognize patterns ranging from lipopolysaccharide (LPS) to nucleic acids such as viral RNA and CpG DNA [184,185,186,187,188]. The subtype TLR4 forms a multi-receptor complex on the surface of a cell with several other proteins and adaptors. LPS, TLR4’s primary agonist, is comprised of 3 primary structure domains: (1) the lipid A tail, (2) the core oligosaccharide, and (3) the O antigen (Fig. 1) [189]. The hydrophobic lipid A tail is a motif that is recapitulated in not only pathogens, but DAMPs produced in vivo, and the backbone of fatty acids consumed in the western diet [34, 190, 191]. TLR4 is expressed on cells in both the CNS and periphery including microglia [192], astrocytes [193], neurons [194], macrophages [195, 196], endothelial cells [197], fibroblasts [198] and more. Since TLR4 is expressed on a variety of cells with specific biological functions, considerations must be made on a per cell basis when interpreting the effects of TLR4 signaling in disease pathology.

Evidence suggests that in the DRG and trigeminal ganglia (TG), TLR4 and TRPV1 are functionally coupled and TLR4 can directly modulate activity of TRPV1 [199, 200]. Infusion of LPS on dissociated TG and DRG neurons potentiated TRPV1 agonist responses such as calcium influx and CGRP release, both of which are not directly linked to TLR4 signaling cascades [201]. In TLR4 knockout mice, TRPV1 agonist-induced pain is significantly reduced [199]. Moreover, TLR4 possesses the ability to modulate sodium channel (Nav) activity—an essential component of nociceptive transmission. Upon activation, TLR4 initiates an intracellular signaling cascade that activates NF-κB which interacts with pain sensitive Navs on nociceptors within minutes of activation to increase Nav currents and decrease recovery time from inactivation, which enhance neuronal excitability and pain signaling [202]. Taken together, these studies reveal a unique interaction between TLR4 and TRPV1 as well as TLR4 and Navs, which enhance the context by which we understand nociceptor physiology.

Specific modulation of pain by TLR4

Evidence has mounted that TLR4 is a critical mediator of the nervous and immune system’s inflammatory response during pain [91, 202,203,204]. TLR4 gene expression is increased in the spinal cord when rats are injected with intraplantar CFA [205]. Subsequent studies have verified this by showing an increase in microglia reactivity in the dorsal spinal cord following intraplantar CFA injection [206]. Another study also demonstrated the involvement of TLR4 in acute inflammatory pain by utilizing intrathecal injections of high mobility group box 1 (HMGB1), a TLR4 agonist, which enhanced activation of microglia in a TLR4-dependent manner subsequently producing hypersensitivity in mice [207].

TLR4 participates in several inflammatory conditions that have previously been found to differ by sex. For example, rheumatoid arthritis, has a more pronounced effect in women [208]. In a rodent model of arthritis, it was revealed that while both females and males demonstrate allodynia after inflammation, males continue to have tactile allodynia after joint inflammation has resolved [208]. However, global deletion of TLR4 in both sexes attenuated the post-inflammatory allodynia [208]. In a similar model, a global deletion of TLR4 attenuated the post-inflammatory allodynia in males, whereas an intrathecal injection of a TLR4 antagonist given during the inflammatory phase fully reversed the pain-like behavior associated with arthritis in males [209]. These findings suggest that TLR4 signaling alone can sustain the inflammation seen in arthritis and contributes to sex differences in the manifestation of the disorder.

In addition to inflammatory pain, TLR4 has also been shown to be involved in chemotherapy-induced peripheral neuropathy (CIPN). CIPN is one of the major dose-limiting side effects of cancer treatments. Paclitaxel, a taxane-based chemotherapeutic, engages similar inflammatory intracellular signaling pathways as the classical TLR4 ligand, LPS [210]. This is evident by paclitaxel utilizing the same accessory protein required for LPS signal transduction and upregulating inflammatory cytokines such as IL-1β [211]. Paclitaxel treatment upregulates TLR4 expression in the DRG and spinal cord, suggesting its connection with the development of pain [212], and blocking TLR4 signaling reverses this pain phenotype [213].

TLR4 plays a significant role in opioid-induced hyperalgesia [214, 215]. While this seems paradoxical, evidence suggests that µ-opioids can lead to increased pain sensitivity [216]. It is believed that certain opioids can bind to TLR4, which facilitates inflammatory signaling [217]. By acting as TLR4 ligands, traditional opioid receptor agonists induce a pain phenotype or enhance sensitivity [218].

Sex differences in TLR4-mediated pain development and allodynia

Over the last decade, TLR4 has made its way into the spotlight of pain research. However, its exact sex- and cell-specific role in mediating chronic pain development and maintenance have been controversial. One of the first studies to implicate TLR4 signaling in neuropathic pain development sought to characterize the functional role of microglia TLR4 in response to neuropathic injury [219]. They found that TLR4 whole body null mice had an attenuated spinal microglial inflammatory response, but also a reduced nociceptive behavioral phenotype after nerve injury. Subsequent studies have corroborated these findings by pharmacologically inhibiting spinal TLR4 activation [220, 221]. While these studies do provide convincing evidence that suggests immune-mediated TLR4 signaling contributes to neuropathic pain development, sex was not considered as a variable. It was not until a few years later that the sex-specific contribution of TLR4 in both inflammatory and neuropathic pain was addressed [183]. It was found that intrathecal administration of LPS induced a robust pain phenotype in male but not female mice, and antagonism of spinal TLR4 reversed this effect in only males. Moreover, neuronal injury induced neuropathic pain behaviors in both male and female mice, but only male mice showed transient attenuation of pain behaviors following intrathecal TLR4 antagonist injection. Some data suggest that the role of TLR4 in inflammatory and neuropathic pain development is reliant on the timing of TLR4 modulation [209, 222]. While another study demonstrated that intrathecal LPS facilitated mechanical hypersensitivity development in both male and female mice, seemingly contrasting findings of TLR4 inhibition were noticed [183]. Inhibition of spinal TLR4 signaling by intracellular antagonist, TAK-242, prevented development of allodynia, but could not reverse established allodynia in males [223]. In accordance with the previously discussed study, another group demonstrated that TLR4 whole-body null females do not exhibit a reduction in mechanical hypersensitivity like their male counterparts after neuropathic injury [224]. This was one of the first studies to highlight that TLR4 either directly or indirectly influences expression of neuronal injury markers in the periphery. While these results seem contradictory, they may be explained by mouse strain differences used [225,226,227,228] and serotype and dose of LPS [229,230,231].

To address discrepancies in the temporal and cell-specific role of TLR4 in chronic pain, a recent study used microglia-specific TLR4 conditional knockout mice, in which a tamoxifen-dependent Cx3CR1-CreERT2−EYFP mouse line was used [232]. They found that systemic antagonism of TLR4 is effective at improving chronic pain outcomes in a tibial fracture model of complex regional pain syndrome in both sexes when administered at the time of injury as opposed to when pain is already established. Their data showed removing microglial TLR4 after the injury resulted in partial improvement in allodynia of both sexes, suggesting that microglial TLR4 plays less of a role in the maintenance of chronic pain states [232]. Interestingly, their investigation showed a female-specific reliance on peripheral TLR4 expressing myeloid cells to trigger chronic pain. In a different context of pain, recent evidence uses a novel null-reactivatable approach that microglial TLR4 is crucial in mediating female-specific acute mechanical allodynia following ethanol exposure [233]. While microglia-restricted TLR4 expression is sufficient to induce allodynia in females, its role in males is less pronounced, underscoring the potential contributions of TLR4 on tissue macrophages in shaping pain responses in both sexes [233]. Differences in microglial pro-inflammatory signatures and nociceptive signaling demonstrate nuanced neuroimmune interactions of central and peripheral TLR4 signaling. This suggests that TLR4 signaling cascades may be different between sexes and temporal dynamics may influence cell-specific actions of TLR4 in the transition from acute to chronic pain.

Other important considerations in studying the effects of TLR4 in chronic pain are the models and inflammatory mediators used. HMGB1 is a highly dynamic nuclear chromatin-binding protein that orchestrates key biological events such as sterile inflammation and chemoattraction based on redox biology [234]. HMGB1 can signal through multiple receptors including TLR4 [235, 236]. Passive secretion of HMGB1 results from cell death, injury, autophagy, and necrosis [237,238,239]. Intracellular HMGB1 exists in its all-thiol form (at-HMGB1) and can be oxidized to the disulfide form (ds-HMGB1) when reactive oxygen species present in the extracellular matrix facilitate disulfide bridge formation between cysteine residues 23 and 45 [240]. Ds-HMGB1 functions as an inflammatory cytokine that induces preferential activation of TLR4 [241]. HMGB1 is upregulated in sensory neurons in the DRG, as well as peripheral immune cells and glia in response to neuropathic injury [151]. Upregulation of HMGB1 was also associated with a transition from nuclear to cytosolic localization, which is indicative of active release [242]. Inhibition of HMGB1 signaling by a neutralizing antibody prevents mechanical allodynia development and cytosolic localization of the protein, suggesting that peripheral HMGB1 signaling plays a significant role in the development of neuropathic pain [243]. Moreover, only male rats were used in these experiments. Another study demonstrated that administration of ds-HMGB1 increased excitability of dissociated DRG neurons and treatment with glycyrrhizin, an HMGB1 inhibitor, reversed the HMGB1-dependent increase in neuronal excitability. Moreover, systemic inhibition of HMGB1 signaling reversed mechanical allodynia resulting from nerve injury [244]. A more recent study highlighted the effects of central HMGB1-TLR4 signaling in arthritis-like pain [245]. Intrathecal administration of ds-HMGB1 induced pain-like behaviors in mice of both sexes and is TLR4-dependent. Moreover, pharmacological inhibition of spinal HMGB1 signaling reverses arthritis pain. This study provides additional evidence to support the role of HMGB1 in pain, but it was also one of the first to directly assess sex differences in HMGB1-TLR4 signaling [245]. Although, these findings do not recapitulate the Sorge et al., 2011 study that found robust sex differences in spinal TLR4 signaling suggesting mechanistic differences in the specific ligands, signaling pathways, and cell types responsible for neuropathic pain development between males and females [183]. The involvement of sex- and cell-specific HMGB1 signaling in the periphery has not been extensively studied until recently.

A recent study found that inhibition of TLR4 signaling in peripheral macrophages attenuated ds-HMGB1-induced pain behaviors in male but not female mice, suggesting that the cell and tissue types responsible for mediating peripheral pain mechanisms differ between sexes as well [246]. In the same study, using conditional DRG nociceptor TLR4 knockout animals demonstrated that HMGB1 mediates arthritis pain through TLR4 signaling in both sexes. Another study confirmed these findings by using a similar genetic and pain model [247]. Moreover, by conditional deletion of HMGB1 from the whole DRG neuron population, they confirm that HMGB1 is also responsible for neuropathic pain development during neuronal injury in females. The specific receptors for HMGB1, neuronal subtypes, and male-specific mechanisms were not addressed [247]. Therefore, studies that evaluate the effectiveness of inhibiting both TLR4 and HMGB1 signaling will provide insight on the sex- and cell-specific mechanisms of chronic pain and provides a prime example of a mediator of sex-specific differences in neuroimmune interactions.

Hormonally driven sex differences in neuroimmune communications, pain, and physiology

It is now widely appreciated that the nervous and immune systems communicate with one another to coordinate their dynamic responses. It is also apparent that the endocrine system and hormones also play a role in these actions [248]. The interaction between the nervous, endocrine, and immune systems is bidirectional, indicating that crosstalk between these systems is necessary to not only maintain homeostasis, but also to respond appropriately to tissue injury and infection [36, 248]. While the scientific community has traditionally focused on male-biased research, it is making significant strides to assess differences in nervous and immune system communication between males and females. However, there are still unanswered questions and mechanisms that remain unclear. For instance, TRPV1 activity is well-known for promoting pain and inflammation and can be differentially regulated by different sex steroids. Recent studies found that estradiol levels influence TRPV1 expression in females [249,250,251], positively modulating its expression to promote pain. However, studies are still inconclusive on whether testosterone modulates TPRV1 expression or activity in humans [252,253,254]. Studies are still quite sparse regarding sex differences in pain, and thus more studies are needed to unravel the influence of hormones on sex-specific biomarkers driving these dimorphisms.

Accounting for differences between males and females is important as some conditions have a greater prevalence in females compared to males. Moreover, studies confirm that neuroendocrine interactions with the immune system play a role in the pathogenesis of these conditions [255]. For example, males have lower susceptibility to autoimmune disorders compared to females due to testosterone’s protective immunomodulatory function [255, 256]. Studies show that a disproportionate number of females are impacted by autoimmune diseases, chronic pain disorders, and the lack of effective therapeutics [255, 257, 258]. Thus, understanding the sex- and cell-specific differences in neuroimmune modulation is necessary to appropriately design and execute research targeted at effective therapeutic development.

Testosterone

Androgens have been found to mediate sex differences in neuroimmune interactions and within the last decade, numerous studies have highlighted testosterone as a mediator of sex differences in immunity and pain development [259,260,261]. Studies have shown that testosterone receptors, also called androgen receptors (AR), modulate immune and pain outcomes in gonadectomized mice by promoting µ-opioid and cannabinoid receptor activity in the presence of high levels of testosterone [262,263,264]. Another study using a model of chronic inflammatory pain in rats demonstrated that testosterone inhibits TRPV1 expression [265]. Although testosterone serves as the primary sex hormone in males, dysregulation in testosterone production has clinical implications in both sexes. In females, hyperandrogenism is associated with polycystic ovary syndrome (PCOS) [266]. PCOS is an endocrine disorder affecting females of reproductive age and has several associated comorbidities including insulin resistance and cardiovascular disease [267]. Further, in females with PCOS, increased levels of androgens correlate with increased cytokine secretion and disrupts the normal balance of the immune system by stimulating some immune cells and inhibiting others, which can produce a state of chronic, low-grade inflammation [268]. It is especially important that normal-range testosterone levels be maintained in both sexes considering the potential risks associated with immune cardiovascular diseases. A previous study found that administration of testosterone in males with androgen deficiency led to decreased levels of pro-inflammatory cytokines IL-1β and TNF-α and an increase in the anti-inflammatory cytokine IL-10 [269]. Testosterone has also been shown to act directly on CD4+ T-cells to increase IL-10 production [256]. Furthermore, research has indicated that testosterone replacement therapy may favorably shift cytokine balance and reduce total cholesterol in males, potentially improving cardiovascular disease risk [269]. Testosterone has also been shown to protect against the development of widespread muscle pain, a hallmark of the female-dominated condition fibromyalgia, in an activity-induced pain model [259]. Hence, more studies unraveling in-depth mechanisms of testosterone in neuroimmune communication are crucial to addressing pain disparities between males and females.

Estrogens and progesterone

Estrogens are produced mainly by the ovaries in females and to a lesser extent, the testes in males [270]. Estrogens are involved in a variety of physiological processes including the resolution of inflammation and may be a promising therapeutic for promoting anti-inflammation [271]. Fluctuations in estrogens and progesterone during the estrus cycle of female rodents alter the circadian rhythm of both physiology and behavior [272]. The two subtypes of estrogen receptors (ER), ER-α and ER-β, bind estrogens and differentially affect inflammation and pain modulation [273]. ER-α has been shown across multiple studies to modulate pain [275,276,277]. Estradiol (E2), the most prominent estrogen in the human body, signaling increases NMDA receptor activity while ER-α antagonists administered in the spinal cord increase excitatory postsynaptic currents, which may increase pronociceptive signaling [277, 278]. Further, both male and female ER-α knockout mice show small increases in nociceptive behaviors, suggesting that ER-α signaling inhibits pain transmission [276]. There is evidence, however, that ER-α and ER-β signaling may be pronociceptive, as another study found that knockout of either receptor significantly increases elevated responses to mechanical stimuli in female mice [275]. In most neurons with both receptor subtypes co-expressed, high estrogen levels can result in the dimerization of ER-α and ER-β, which suppresses ER-α signaling [280,281,282]. In a study looking at how estrogens affect µ-opioid receptor levels and activation with a pain stressor in females, low estrogen levels corresponded with increased pain perception and a reduction in the endogenous opioid neurotransmission function [282]. E2 has been found to mitigate the negative effects of glucocorticoids in cognitive processes [283]. Furthermore, E2 positively activates TRPV1 channels at higher concentrations [249, 251, 284, 285]. The interactions between neurotransmitters like serotonin and estrogens have been linked to female-specific differences in immune modulation and pain perception in conditions like irritable bowel syndrome and fibromyalgia [286]. Serotonin is considered an inflammatory mediator and increases pronociceptive signaling. ER activity can increase serotonin’s pronociceptive activity by enhancing postsynaptic responsiveness and increasing serotonin synthesis in the brain [287].

Although both estrogens and progesterone play a role in modulating pain and inflammation, progesterone’s specific mechanisms are not as well-known as that of estrogen. Progesterone is secreted by the corpus luteum where it has downstream implications in reproductive and immune processes, especially during pregnancy [288]. While progesterone has been extensively studied in relation to female reproductive processes, its role in males is not yet fully understood [289]. Immune cells, including macrophages, NK cells, dendritic cells, and T-cells, express progesterone receptors [290]. Progesterone suppresses macrophage and dendritic cell activation, alters T-cell distribution and activity, and causes apoptosis of NK cells expressing progesterone receptors [290, 291]. Via modulation of transcription factors, progesterone decreases inflammation by inhibiting pro-inflammatory cytokine production and increasing the production of anti-inflammatory cytokines [293,294,295]. A study investigating progesterone’s anti-inflammatory effects in LPS-stimulated microglia found that progesterone inhibits NF-kB activation [295]. Another study reports that peaks in progesterone and testosterone are associated with lower reported pain severity in females with fibromyalgia, suggesting both sex hormones possess a protective role in modulating fibromyalgia pain [21].

Medications containing hormones, such as oral contraceptive pills containing both estrogens and progestins, can also influence pain and contribute to pain states. These contraceptive pills are the most commonly prescribed contraceptive pills in the United States, however, individuals who experience migraines are not recommended to take them due to the increased risk of migraines [296]. Fluctuations in hormone levels during the menstrual cycle is also believed to play a role in the pathogenesis of migraine attacks and signaling of these hormones may explain the greater prevalence of migraine in females [297].

Prolactin

In addition to estrogens and progesterone, prolactin, a hormone produced by the pituitary gland and lymphocytes, is also implicated in driving neuroimmune interactions in pain [248, 298]. Prolactin secretion is regulated by cytokines, but can also function as a cytokine itself, with the prolactin receptor (PRLRs) being included in the cytokine receptor super family [299, 300]. Prolactin stimulates PRLRs of B-cells, macrophages, and T-cells to induce immune responses [301]. Prolactin plays a critical role in the complex sex-specific mechanisms underlying migraine [302, 303]. Reports show a direct correlation between high prolactin levels and the prevalence of chronic migraine, with increased risk of migraines in patients with hyperprolactinemia [306,307,308,309]. Additionally, a preclinical study showed dural administration of prolactin resulted in female-specific migraine-like behavior in both cycling and ovariectomized rodents [302, 303, 308]. Overexpression of prolactin and its receptors are causal factors in the development of autoimmunity and pain. For example, hyperprolactinemia plays a role in the clinical manifestations of some autoimmune diseases including multiple sclerosis and Sjogren’s syndrome [309]. Furthermore, using both local and spinal inflammatory pain models, a previous study found that PRLR contributes to hypersensitivity in a female-specific manner [310]. Additionally, PRLR has been found to contribute to sexual dimorphic differences in pain during inflammatory processes in neuronal and non-neuronal cells [310, 311]. In a study that used a mouse line with ablated PRLR in sensory neurons, they found that PRLR in sensory neurons is critical for transitioning pain from the acute to chronic state in females only [312]. Additionally, PRLR expression is higher in neuronal fibers and dural mast cells in female mice while male mice showed little to no expression, which may contribute to increased prevalence of migraines in females [308].

Nuclear receptors: peroxisome proliferator-activated receptors (PPARs)

While hormones and nuclear receptor ligands can contribute to sex differences in neuroimmune communications, they can also serve as modulators of other components that lead to sexual dimorphisms. Peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptor proteins comprised of three different subtypes (PPARα, PPARγ, and PPARβ/δ). PPARs are involved in glucose metabolism, cell survival, lipid detection, and immunity [313]. While PPARs are expressed in a variety of tissues, the expression of subtypes α and γ in T-cells has been shown to be sexually dimorphic in its contribution to pain and inflammation [314]. PPARα is found in macrophages and lymphocytes where it plays a role in suppressing inflammatory activation [315]. Elevated levels of testosterone in males increase the expression of PPARα, which inhibits IFN-γ, thus suppressing inflammation [316]. A study also found that administration of a PPARα agonist alleviated pain specifically in male mice and this was reversed by eliminating testosterone [166]. Conversely, PPARγ inhibits macrophages and T-cell proliferation and plays a key role in regulatory T-cell activity. Elevated levels of estrogen in females increase PPARγ expression, which inhibits NF-κB and T-cells and suppresses inflammation. Administration of a PPARγ agonist alleviated pain specifically in female mice, and this was reversed by testosterone [166]. Taken together, PPARs and hormones highlight additional mechanisms in which elements of the immune system drive sex differences in pain and inflammation.

Sex differences in pain and peripheral inflammation in the clinic

The neuroimmune mechanisms underlying sex differences and their impact on chronic pain conditions remains a critical research question for clinical translation. Although animal models provide insight into mechanisms underlying pain, neuroimmune communication, and pain resolution, lack of our understanding of nuances between humans and non-human models slows progress. For example, preclinical rodent models suggest females are less responsive to pain therapeutics, such as low efficacy opioids, however, clinical studies indicate the opposite effect in humans, with opioids generally being more effective and potent in females than males [317, 318]. The severity of the ongoing opioid epidemic is evident in the reported 4 and 5.8-fold increase in overdoses since 1999 in males and females respectively [319, 320]. These increases in overdoses make evident the importance of not only advancing pain research, but also better evaluating the differences across sexes.

Additionally, although existing preclinical and clinical research provides some insight into the sex differences in pain in patients, further investigation is needed to fully elucidate the differences in physiology between males and females to develop and prescribe more personalized, effective pain treatments for patients. Given the higher prevalence of chronic pain conditions in females than in males [321], pain research has honed in on efforts to characterize potential sex differences in actual pain sensitivity and mechanisms [12, 322].

Sex differences in endogenous descending pain modulation have been observed in the form of diffuse noxious inhibitory controls (DNIC). DNIC dampens pain signaling in the dorsal horn of the spinal cord via inhibitory neurotransmitters. Research has found that DNIC is more pronounced in males compared to females [323]. However, as with most clinical data, this evidence is inconsistent due to additional psychological factors rendering DNIC sex differences insignificant [324]. In the clinic, females typically have less favorable outcomes regarding therapeutic intervention than males for pain treatment. In the case of musculoskeletal pain, females are more likely to develop chronic knee pain following total knee arthroplasty and twice at risk of experiencing no clinically important improvement in the case of knee osteoarthritis and widespread pain [325].

The sex differences in pain observed clinically are likely driven by many different cell types and their interactions. For example, immune cells can induce sexually dimorphic pain states through inflammation. In patients with periodontitis, females are more likely to have lower pain thresholds and more persistent pain with macrophage activity likely driving these differences [326]. Furthermore, this study found that genes associated with either promotion or inhibition of nociception differed between sexes [326]. Although the exact immune cells that drive many pain states are still under investigation, sex differences in the levels of pro-inflammatory mediators that drive pain and are involved in the recruitment of immune cells have been reported. For example, in inflammatory dental pain, women show a greater expression in the purinergic receptor, P2X 3, and CD39, a marker for chronic ATP release, than men, which was indicative of greater pain in women than men [327].Specific immune cells have also been implicated in osteoarthritis in females and males, with osteoarthritis females having a higher percentage of CD4+ T cells and macrophages, but a lower percentage of monocytes and CD8+ T cells in their synovial fluid compared to osteoarthritis males [328]. These findings indicate that differences in inflammation-related physiology such as pain can be attributed to sex differences.

Macrophage polarization in asthma and headache related pain

Male and female macrophages differ in propensity to polarize towards pro- and anti-inflammatory states [329, 330]. There is a sex-dependent functional state of immune cells in asthmatic patients, with macrophages and monocytes exhibiting a greater anti-inflammatory M2 phenotype in females [331]. Asthma is also a comorbidity of cluster headaches that is more prevalent in females [332]. There may be a bidirectional etiology where cluster headaches cause further alterations to monocytes, and asthma’s underlying alterations of the immune state may be a contributing factor to cluster headache development [332]. For example, asthma-associated inflammation triggers vascular neuroinflammation to induce headaches, and asthma treatment options are also capable of inducing headaches [333]. While females and males both experience retro-orbital pain with cluster headaches, females disproportionately suffer from coexisting jaw, ear, and cheek pain, as well as migraine-like symptoms such as nausea [332]. Furthermore, sex differences exist in treatment for headache pain, as females respond more to lidocaine and CGRP antagonist, whereas males respond more to sumatriptan [332, 336,337,338].

Implications of adiposity and pain

Adiposity serves as a key consideration in the evaluation of risk for obesity, and its sexually dimorphic character contributes to sex disparities in clinical settings, as males and females have uniquely different adipose distributions [337]. Adiposity influences immune responses and has both a direct physical and physiological role in the development of chronic pain. Female adipose distribution is characterized by higher femoral deposition, along with increased levels of inflammatory C-reactive protein (CRP). Moreover, females exhibit higher levels of IL-6 and leptin, a hormone released by adipose tissue, which is implicated in exacerbating chronic pain severity in female patients [338]. Differential plasma levels of inflammatory markers, such as CRP, in females are specifically due to greater accumulation of subcutaneous fat compared to male counterparts [339]. In the case of diet-induced obesity, males and females express distinct inflammatory responses, with males having higher levels of macrophage accumulation in subcutaneous white adipose tissue. This macrophage accumulation is linked to elevated fatty acid species in these tissues, in which their storage and release causes this male-specific inflammation [340]. Preclinical models have demonstrated higher levels of adiposity directly correlate to increased TLR4 expression to mediate pro-inflammatory responses in tissue highlighting another crucial role for TLR4 in the attribution of pain phenotypes in a sex-dependent manner [343,344,345]. This translates to the clinic where researchers discovered obese patients have high adipocyte TLR4 expression from subcutaneous abdominal fat [344], suggesting TLR4 may play a role in adiposity-related regulation of immune responses and chronic pain.

Postsurgical recovery and pain

Overall, sex differences in the development of postoperative chronic pain have been identified across many surgery types, with females more frequently reporting severe pain levels one day postoperatively and higher pain scores across the recovery period [345]. Recent studies focused on cardiac surgical outcomes have reported sex differences in postsurgical pain and recovery. In a one year postoperative study, biological females presented with higher levels of postoperative pain than males across time [346], while males had lower mortality rates than females [347, 348]. This indicates that differences in surgical strategy may influence sex differences in recovery outcomes. In total knee arthroplasty procedures, one study found that females experienced more acute postoperative pain, while no sex differences were observed six-weeks postoperatively [349]. Interestingly, this alludes to how biologically, males and females utilize recovery strategies to end up back to homeostasis.

Sex hormone modulation of nociception

As previously stated, sex hormones and their influence are a culprit behind sexually dimorphic nociceptive responses. However, recent preclinical evidence suggests that transient shifts in hormones like estrogen during the estrus cycle in rodents may not be directly responsible for these nociceptive responses, suggesting estrogen level fluctuations during the menstrual cycle may not be directly responsible for nociceptive responses in humans [35, 176, 350]. It is possible that this dichotomy in sex-based pain may be hardwired at or right before birth and modulated to a lesser degree by the fluctuation of hormones during adulthood. As evidence, a recent clinical study in pre-and full-term infants discovered that widespread nociceptive events measured by electroencephalography are different between sexes [351]. Interestingly, no differences were found in response to innocuous touch, indicating that the signaling pathways between the sensory and nociceptive systems are not mutually exclusive.

Sex differences in inflammatory factors in osteoarthritis

Many studies have shown that pain in osteoarthritis influences changes in gait [352], and gait changes contribute to pain in a bidirectional manner. Gait differences exist between sexes both pre- and post-surgically in patients with osteoarthritis, indicating that sex differences exist both in inflammatory osteoarthritis development as well as in recovery from associated surgical intervention [353]. Research in osteoarthritis development has gained traction in recent years, taking into consideration that this condition is more prevalent and painful in females [356,357,358]. Osteoarthritis has been labeled as a condition associated with less inflammation than other arthritis subtypes [356], however, more recent clinical studies have reported sex-differences in the immune response [356,357,358]. These sex differences have been observed in both the development and in persistent pain in osteoarthritis, however, the exact mechanisms have yet to be fully elucidated [356,357,358]. Interestingly, females with osteoarthritis show higher levels of IFN-γ, IL-3, IL-8, IL-18, and TNF-β, and other factors that stimulate macrophages compared to osteoarthritis men [357, 358]. Conversely, others report that IL-8 has been found to be higher in males with osteoarthritis [359]. In addition to sex differences in the levels of inflammatory factors in osteoarthritis, these factors may drive pain differently in females and males. For example, IL-6 levels are correlated with increased pain in females and decreased pain in males, IL-1β and IL-8 levels correlate with increased pain in males and decreased pain in females, which further demonstrates the need for sex specific treatments for osteoarthritis [360]. Collectively, these instances of sexual dimorphisms within the clinic emphasize the importance of understanding the influence of sex on neuroimmune interactions and their relation to pain and medicine.

Conclusions and future perspectives

The neuroimmune “conversation” is implicated in pain physiology, whether through signaling between immune cells and sensory neurons or immune components expressed by neurons. A prime example of bidirectional neuroimmune communication in pain physiology is neuron-microglia crosstalk, in which neurons activate microglia through glutamate or ATP release and activated microglial actions modulate neuronal excitability [142, 144, 145]. Depending on the pain state and the model used, the cell types that drive sexual dimorphisms in pain differ. Sex differences in pain are further influenced by condition and the activity of different cell types, such as microglia in the CNS driving chronic pain development in a sex-specific manner, and by the neuronal expression of prototypic and immune receptors [35]. For example, neuroimmune interactions facilitated by sensory neurons via TLR4 and NLRP3 lead to greater pain sensitivity in females compared to males [151, 152]. The endocrine system and hormones further drive sex differences in neuroimmune communication and pain. Estrogen and progesterone serve as modulators of nuclear hormone receptors, like PPARs, that lead to sexual dimorphisms in immunity and pain. While there is a disproportionate number of females impacted by autoimmune diseases and chronic pain disorders seen clinically, the mechanisms underlying this disparity are understudied. Clinicians and researchers are recognizing the impact of understanding sexual dimorphisms in neuroimmune communication to treat patients more effectively. With this realization, it is paramount that bench-to-bedside approaches are taken with the consideration of the complexities of the individual systems that influence each other, along with the complexities of the human condition. To date, multiple neuroimmune experimental models and clinical findings highlight the influence of sex on physiology and behavior. Females make up ~ 50% of the population, however, until recently, have been excluded from scientific studies and clinical trials that directly impact their health and well-being. It is imperative that studies at all levels include a reasonable representation of sex groups to power statistical analysis to find meaningful results for both sexes. While achieving such numbers, superimposed on the fact that many studies necessitate including several female groups at various stages of the estrus cycle and hormone level determination, makes studies on sex differences expensive and time consuming. However, exclusion of these factors leaves large gaps in our scientific understanding in how pain is driven differently in females and males. Determining the minimum numbers needed for the appropriate statistics will drive research standards and best practices. Elucidating the role of sex differences in neuroimmune communication and pain is essential for the development of efficacious treatments for inflammatory and painful conditions in both sexes.

Data availability

Not Applicable.

References

  1. Kourounakis P, Selye H. Influence of steroids and stress on toxicity and disposition of tetraethylammonium bromide. J Pharm Sci. 1976;65:12:1838–40. https://doi.org/10.1002/jps.2600651236.

    Article  CAS  PubMed  Google Scholar 

  2. Hadamitzky M, Lückemann L, Pacheco-López G, Schedlowski M. Pavlovian conditioning of immunological and neuroendocrine functions. Physiol Rev. 2020;100:1:357–405. https://doi.org/10.1152/physrev.00033.2018.

    Article  CAS  PubMed  Google Scholar 

  3. Kourounakis P, Szabo S, Selye H. Effect of pregnenolone-16alpha-carbonitrile on the rat liver. Arzneimittelforschung. 1976;26(1):74–5. https://www.ncbi.nlm.nih.gov/pubmed/947184.

    CAS  PubMed  Google Scholar 

  4. Du Ruisseau P, Tache Y, Selye H, Ducharme JR, Collu R. Effects of chronic stress on pituitary hormone release induced by combined hemi-extirpation of the thyroid, adrenal and ovary in rats. Neuroendocrinology. 1977;24:3–4. https://doi.org/10.1159/000122707.

    Article  Google Scholar 

  5. Salas M, Tuchweber B, Kourounakis P, Selye H. Temperature-dependence of stress-induced hepatic autophagy. Experientia. 1977;33:5:612–4. https://doi.org/10.1007/BF01946531.

    Article  CAS  PubMed  Google Scholar 

  6. Cohen N, Ader R. Immunomodulation by classical conditioning. Adv Biochem Psychopharmacol. 1988;44:199–202. https://www.ncbi.nlm.nih.gov/pubmed/3041747.

    CAS  PubMed  Google Scholar 

  7. Ader R, Cohen N. Conditioning of the immune response. Neth J Med. 1991;39:3–4. https://www.ncbi.nlm.nih.gov/pubmed/1791889. :263 – 73.

    Google Scholar 

  8. Cohen N, Moynihan JA, Ader R. Pavlovian conditioning of the immune system. Int Arch Allergy Immunol. 1994;105(2):101–6. https://doi.org/10.1159/000236811.

    Article  CAS  PubMed  Google Scholar 

  9. Raison CL, Miller AH. Is depression an inflammatory disorder? Curr Psychiatry Rep. 2011;13:6. https://doi.org/10.1007/s11920-011-0232-0.

    Article  Google Scholar 

  10. Raison CL, Miller AH. Role of inflammation in depression: implications for phenomenology, pathophysiology and treatment. Mod Trends Pharmacopsychiatry. 2013;28:33–48. https://doi.org/10.1159/000343966.

    Article  CAS  PubMed  Google Scholar 

  11. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:1:22–34. https://doi.org/10.1038/nri.2015.5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Miller LR, Marks C, Becker JB, Hurn PD, Chen WJ, Woodruff T, et al. Considering sex as a biological variable in preclinical research. Faseb J. 2017;31:1:29–34. https://doi.org/10.1096/fj.201600781R.

    Article  CAS  PubMed  Google Scholar 

  13. Alshawaf AJ, Viventi S, Qiu WZ, D’Abaco G, Nayagam B, Erlichster M, et al. Phenotypic and functional characterization of peripheral sensory neurons derived from human embryonic stem cells. Sci Rep. 2018;8:12. https://doi.org/10.1038/s41598-017-19093-0.

    Article  CAS  Google Scholar 

  14. Li CL, Li KC, Wu D, Chen Y, Luo H, Zhao JR, et al. Somatosensory neuron types identified by high-coverage single-cell RNA-sequencing and functional heterogeneity (26, pg 83, 2016). Cell Res. 2016;26:8967. https://doi.org/10.1038/cr.2016.90.

    Article  CAS  Google Scholar 

  15. Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, et al. Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med. 2022;14:632eabj8186. https://doi.org/10.1126/scitranslmed.abj8186.

    Article  CAS  Google Scholar 

  16. Yu H, Usoskin D, Nagi SS, Hu Y, Kupari J, Bouchatta O et al. Single-Soma Deep RNA sequencing of Human DRG Neurons Reveals Novel Molecular and Cellular Mechanisms Underlying Somatosensation. bioRxiv. 2023; https://doi.org/10.1101/2023.03.17.533207

  17. Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci. 2005;6:7. https://doi.org/10.1038/nrn1700.

    Article  CAS  Google Scholar 

  18. Goldberg DS, McGee SJ. Pain as a global public health priority. BMC Public Health. 2011;11:770. https://doi.org/10.1186/1471-2458-11-770.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Dahlhamer J, Lucas J, Zelaya C, Nahin R, Mackey S, DeBar L, et al. Prevalence of Chronic Pain and High-Impact Chronic Pain among adults - United States, 2016. MMWR Morb Mortal Wkly Rep. 2018;67:36:1001–6. https://doi.org/10.15585/mmwr.mm6736a2.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Berkley KJ. Sex differences in pain. Behav Brain Sci. 1997;20:3:371 – 80; discussion 435–513; https://doi.org/10.1017/s0140525x97221485

  21. Schertzinger M, Wesson-Sides K, Parkitny L, Younger J. Daily fluctuations of Progesterone and Testosterone are Associated with Fibromyalgia Pain Severity. J Pain. 2018;19:4:410–7. https://doi.org/10.1016/j.jpain.2017.11.013.

    Article  CAS  PubMed  Google Scholar 

  22. Ruau D, Liu LY, Clark JD, Angst MS, Butte AJ. Sex differences in reported Pain Across 11,000 patients captured in Electronic Medical records. J Pain. 2012;13:3. https://doi.org/10.1016/j.jpain.2011.11.002.

    Article  Google Scholar 

  23. Russell MB, Rasmussen BK, Thorvaldsen P, Olesen J. Prevalence and sex-ratio of the subtypes of migraine. Int J Epidemiol. 1995;24:3:612–8. https://doi.org/10.1093/ije/24.3.612.

    Article  CAS  PubMed  Google Scholar 

  24. Faria V, Erpelding N, Lebel A, Johnson A, Wolff R, Fair D, et al. The migraine brain in transition: girls vs boys. Pain. 2015;156:11:2212–21. https://doi.org/10.1097/j.pain.0000000000000292.

    Article  PubMed  Google Scholar 

  25. Fillingim RB, King CD, Ribeiro-Dasilva MC, Rahim-Williams B, Riley JL. Sex, gender, and Pain: a review of recent clinical and experimental findings. J Pain. 2009;10:5. https://doi.org/10.1016/j.jpain.2008.12.001.

    Article  Google Scholar 

  26. Burrell BD. Comparative biology of pain: what invertebrates can tell us about how nociception works. J Neurophysiol. 2017;117:4:1461–73. https://doi.org/10.1152/jn.00600.2016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Black JA, Nikolajsen L, Kroner K, Jensen TS, Waxman SG. Multiple sodium channel isoforms and mitogen-activated protein kinases are present in painful human neuromas. Ann Neurol. 2008;64:6644–53. https://doi.org/10.1002/ana.21527.

    Article  Google Scholar 

  28. Song MY, Yuan JX. Introduction to TRP channels: structure, function, and regulation. Adv Exp Med Biol. 2010;661:99–108. https://doi.org/10.1007/978-1-60761-500-2_6.

    Article  CAS  PubMed  Google Scholar 

  29. Frederick J, Buck ME, Matson DJ, Cortright DN. Increased TRPA1, TRPM8, and TRPV2 expression in dorsal root ganglia by nerve injury. Biochem Biophys Res Commun. 2007;358:4:1058–64. https://doi.org/10.1016/j.bbrc.2007.05.029.

    Article  CAS  PubMed  Google Scholar 

  30. Patapoutian A, Tate S, Woolf CJ. Transient receptor potential channels: targeting pain at the source. Nat Rev Drug Discov. 2009;8(1):55–68. https://doi.org/10.1038/nrd2757.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yu L, Yang F, Luo H, Liu FY, Han JS, Xing GG, et al. The role of TRPV1 in different subtypes of dorsal root ganglion neurons in rat chronic inflammatory nociception induced by complete Freund’s adjuvant. Mol Pain. 2008;4:61. https://doi.org/10.1186/1744-8069-4-61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Duitama M, Vargas-López V, Casas Z, Albarracin SL, Sutachan JJ, Torres YP. TRP channels role in Pain Associated with neurodegenerative diseases. Front Neurosci. 2020;14:782. https://doi.org/10.3389/fnins.2020.00782.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Boyd JT, LoCoco PM, Furr AR, Bendele MR, Tram M, Li Q, et al. Elevated dietary ω-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions. Nat Metab. 2021;3:6762–73. https://doi.org/10.1038/s42255-021-00410-x.

    Article  CAS  Google Scholar 

  34. Tierney JA, Uong CD, Lenert ME, Williams M, Burton MD. High-fat diet causes mechanical allodynia in the absence of injury or diabetic pathology. Sci Rep. 2022;12:1.

    Article  Google Scholar 

  35. Alexander SN, Jeong HS, Szabo-Pardi TA, Burton MD. Sex-specific differences in alcohol-induced pain sensitization. Neuropharmacology. 2023;225:109354. https://doi.org/10.1016/j.neuropharm.2022.109354.

    Article  CAS  PubMed  Google Scholar 

  36. Pinho-Ribeiro FA, Verri WA, Chiu IM. Nociceptor sensory neuron–Immune interactions in Pain and inflammation. Trends Immunol. 2017;38:1:5–19. https://doi.org/10.1016/j.it.2016.10.001.

    Article  CAS  Google Scholar 

  37. Udit S, Blake K, Chiu IM. Somatosensory and autonomic neuronal regulation of the immune response. Nat Rev Neurosci. 2022;23. https://doi.org/10.1038/s41583-021-00555-4. :3:157 – 71.

  38. Wu M, Song G, Li J, Song Z, Zhao B, Liang L, et al. Innervation of nociceptor neurons in the spleen promotes germinal center responses and humoral immunity. Cell. 2024;187:12:2935–e5119. https://doi.org/10.1016/j.cell.2024.04.027.

    Article  CAS  PubMed  Google Scholar 

  39. Cohen JA, Edwards TN, Liu AW, Hirai T, Jones MR, Wu J, et al. Cutaneous TRPV1(+) neurons trigger protective innate type 17 anticipatory immunity. Cell. 2019;178. https://doi.org/10.1016/j.cell.2019.06.022. 4:919 – 32 e14.

  40. Caputa G, Castoldi A, Pearce EJ. Metabolic adaptations of tissue-resident immune cells. Nat Immunol. 2019;20:7.

    Article  Google Scholar 

  41. Gray JI, Farber DL. Tissue-Resident Immune cells in humans. Annu Rev Immunol. 2022;40:1:195–220. https://doi.org/10.1146/annurev-immunol-093019-112809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cesta MF. Normal structure, function, and histology of mucosa-associated lymphoid tissue. Toxicol Pathol. 2006;34:5:599–608. https://doi.org/10.1080/01926230600865531.

    Article  PubMed  Google Scholar 

  43. Panda SK, Colonna M. Innate lymphoid cells in mucosal immunity. Front Immunol. 2019;10:861. https://doi.org/10.3389/fimmu.2019.00861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Randall TD, Mebius RE. The development and function of mucosal lymphoid tissues: a balancing act with micro-organisms. Mucosal Immunol. 2014;7:3. https://doi.org/10.1038/mi.2014.11.

    Article  CAS  Google Scholar 

  45. Seo GY, Giles DA, Kronenberg M. The role of innate lymphoid cells in response to microbes at mucosal surfaces. Mucosal Immunol. 2020;13:3:399–412. https://doi.org/10.1038/s41385-020-0265-y.

    Article  CAS  Google Scholar 

  46. 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. J Pain. 2016;17:10:1081–94. https://doi.org/10.1016/j.jpain.2016.06.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bove D, Lupoli A, Caccavale S, Piccolo V, Ruocco E. Dermatological and immunological conditions due to nerve lesions. Funct Neurol. 2013;28:2:83–91. https://doi.org/10.11138/FNeur/2013.28.2.083.

    Article  Google Scholar 

  48. Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA, Mucida D. Neuro-immune interactions drive tissue programming in intestinal macrophages. Cell. 2016;164:3. https://doi.org/10.1016/j.cell.2015.12.023.

    Article  CAS  Google Scholar 

  49. Riol-Blanco L, Ordovas-Montanes J, Perro M, Naval E, Thiriot A, Alvarez D, et al. Nociceptive sensory neurons drive interleukin-23-mediated psoriasiform skin inflammation. Nature. 2014;510:7503157–61.

    Article  Google Scholar 

  50. Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation. Nature. 2003;421:6921.

    Article  Google Scholar 

  51. Jentho E, Weis S. DAMPs and Innate Immune Training. Front Immunol. 2021;12:699563. https://doi.org/10.3389/fimmu.2021.699563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Barger SW, Goodwin ME, Porter MM, Beggs ML. Glutamate release from activated microglia requires the oxidative burst and lipid peroxidation. J Neurochem. 2007;101:5:1205–13. https://doi.org/10.1111/j.1471-4159.2007.04487.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, et al. Tumor necrosis Factor-α induces neurotoxicity via Glutamate Release from hemichannels of activated Microglia in an Autocrine Manner. J Biol Chem. 2006;281:30:21362–8. https://doi.org/10.1074/jbc.m600504200.

    Article  CAS  PubMed  Google Scholar 

  54. Takeuchi H, Jin S, Suzuki H, Doi Y, Liang J, Kawanokuchi J, et al. Blockade of microglial glutamate release protects against ischemic brain injury. Exp Neurol. 2008;214(1):144–6. https://doi.org/10.1016/j.expneurol.2008.08.001.

    Article  CAS  PubMed  Google Scholar 

  55. Oishi Y, Manabe I. Macrophages in inflammation, repair and regeneration. Int Immunol. 2018;30:11. https://doi.org/10.1093/intimm/dxy054.

    Article  CAS  Google Scholar 

  56. Rosales C, Uribe-Querol E, Phagocytosis. A fundamental process in immunity. Biomed Res Int. 2017;2017:9042851. https://doi.org/10.1155/2017/9042851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen P, Piao X, Bonaldo P. Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol. 2015;130:605–18.

    Article  CAS  PubMed  Google Scholar 

  58. Lim JY, Choi S-I, Choi G, Hwang SW. Atypical sensors for direct and rapid neuronal detection of bacterial pathogens. Mol Brain. 2016;9:1. https://doi.org/10.1186/s13041-016-0202-x.

    Article  CAS  Google Scholar 

  59. Davies AJ, Kim HW, Gonzalez-Cano R, Choi J, Back SK, Roh SE, et al. Natural killer cells degenerate intact sensory afferents following nerve Injury. Cell. 2019;176. https://doi.org/10.1016/j.cell.2018.12.022. 4:716 – 28.e18.

  60. Ristoiu V. Contribution of macrophages to peripheral neuropathic pain pathogenesis. Life Sci. 2013;93:23870–81.

    Article  Google Scholar 

  61. Zhang F, Miao Y, Liu Q, Li S, He J. Changes of pro-inflammatory and anti-inflammatory macrophages after peripheral nerve injury. RSC Adv. 2020;10:64:38767–73. https://doi.org/10.1039/d0ra06607a.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Davies AJ, Rinaldi S, Costigan M, Oh SB. Cytotoxic immunity in peripheral nerve injury and pain. Front NeuroSci. 2020;14:142.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Walker FR, Beynon SB, Jones KA, Zhao Z, Kongsui R, Cairns M, et al. Dynamic structural remodelling of microglia in health and disease: a review of the models, the signals and the mechanisms. Brain Behav Immun. 2014;37:1–14. https://doi.org/10.1016/j.bbi.2013.12.010.

    Article  CAS  PubMed  Google Scholar 

  64. Tian D-S, Peng J, Murugan M, Feng L-J, Liu J-L, Eyo UB, et al. Chemokine CCL2–CCR2 signaling induces neuronal cell death via STAT3 activation and IL-1β production after Status Epilepticus. J Neurosci. 2017;37:33:7878–92. https://doi.org/10.1523/jneurosci.0315-17.2017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Howe CL, LaFrance-Corey RG, Goddery EN, Johnson RK, Mirchia K. Neuronal CCL2 expression drives inflammatory monocyte infiltration into the brain during acute virus infection. J Neuroinflammation. 2017;14:1238. https://doi.org/10.1186/s12974-017-1015-2.

    Article  CAS  Google Scholar 

  66. Baral P, Udit S, Chiu IM. Pain and immunity: implications for host defence. Nat Rev Immunol. 2019;19:7. https://doi.org/10.1038/s41577-019-0147-2.

    Article  CAS  Google Scholar 

  67. Zhang JM, An J. Cytokines, inflammation, and pain. Int Anesthesiol Clin. 2007;45:2:27–37. https://doi.org/10.1097/AIA.0b013e318034194e.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Tecchio C, Micheletti A, Cassatella MA. Neutrophil-derived cytokines: facts beyond expression. Front Immunol. 2014;5:508. https://doi.org/10.3389/fimmu.2014.00508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ozaktay AC, Kallakuri S, Takebayashi T, Cavanaugh JM, Asik I, DeLeo JA, et al. Effects of interleukin-1 beta, interleukin-6, and tumor necrosis factor on sensitivity of dorsal root ganglion and peripheral receptive fields in rats. Eur Spine J. 2006;15:10:1529–37. https://doi.org/10.1007/s00586-005-0058-8.

    Article  PubMed  Google Scholar 

  70. Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, et al. Nociceptors are interleukin-1beta sensors. J Neurosci. 2008;28:52:14062–73. https://doi.org/10.1523/jneurosci.3795-08.2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lynch AM, Walsh C, Delaney A, Nolan Y, Campbell VA, Lynch MA. Lipopolysaccharide-induced increase in signalling in hippocampus is abrogated by IL-10–a role for IL-1 beta? J Neurochem. 2004;88:3. https://doi.org/10.1046/j.1471-4159.2003.02157.x.

    Article  CAS  Google Scholar 

  72. Vitkovic L, Maeda S, Sternberg E. Anti-inflammatory cytokines: expression and action in the brain. Neuroimmunomodulation. 2001;9:6:295–312. ://000176284300001.

    Article  CAS  PubMed  Google Scholar 

  73. Uçeyler N, Schäfers M, Sommer C. Mode of action of cytokines on nociceptive neurons. Exp Brain Res. 2009;196:1:67–78. https://doi.org/10.1007/s00221-009-1755-z.

    Article  CAS  PubMed  Google Scholar 

  74. Gray JI, Farber DL. Tissue-Resident Immune cells in humans. Annu Rev Immunol. 2022;40:195–220. https://doi.org/10.1146/annurev-immunol-093019-112809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022;185:16. https://doi.org/10.1016/j.cell.2022.06.018.

    Article  CAS  Google Scholar 

  76. Segura E, Kapp E, Gupta N, Wong J, Lim J, Ji H, et al. Differential expression of pathogen-recognition molecules between dendritic cell subsets revealed by plasma membrane proteomic analysis. Mol Immunol. 2010;47:9:1765–73. https://doi.org/10.1016/j.molimm.2010.02.028.

    Article  CAS  PubMed  Google Scholar 

  77. Busa R, Bulati M, Badami E, Zito G, Maresca DC, Conaldi PG, et al. Tissue-Resident Innate Immune Cell-based therapy: a cornerstone of immunotherapy strategies for Cancer Treatment. Front Cell Dev Biol. 2022;10:24. https://doi.org/10.3389/fcell.2022.907572.

    Article  Google Scholar 

  78. Jang JH, Clark DJ, Li X, Yorek MS, Usachev YM, Brennan TJ. Nociceptive sensitization by complement C5a and C3a in mouse. Pain. 2010;148:2. https://doi.org/10.1016/j.pain.2009.11.021.

    Article  CAS  Google Scholar 

  79. Ting E, Guerrero AT, Cunha TM, Verri WA Jr., Taylor SM, Woodruff TM, et al. Role of complement C5a in mechanical inflammatory hypernociception: potential use of C5a receptor antagonists to control inflammatory pain. Br J Pharmacol. 2008;153:5. https://doi.org/10.1038/sj.bjp.0707640.

    Article  CAS  Google Scholar 

  80. Pekna M, Pekny M. The complement system: a powerful modulator and effector of astrocyte function in the healthy and diseased Central Nervous System. Cells. 2021;10:71812. https://doi.org/10.3390/cells10071812.

    Article  CAS  Google Scholar 

  81. Ziabska K, Ziemka-Nalecz M, Pawelec P, Sypecka J, Zalewska T. Aberrant complement System Activation in Neurological disorders. Int J Mol Sci. 2021;22:928. https://doi.org/10.3390/ijms22094675.

    Article  CAS  Google Scholar 

  82. Zhou W. The new face of anaphylatoxins in immune regulation. Immunobiology. 2012;217:2225–34.

    Article  Google Scholar 

  83. Alawieh A, Elvington A, Zhu H, Yu J, Kindy MS, Atkinson C, et al. Modulation of post-stroke degenerative and regenerative processes and subacute protection by site-targeted inhibition of the alternative pathway of complement. J Neuroinflamm. 2015;12:1. https://doi.org/10.1186/s12974-015-0464-8.

    Article  CAS  Google Scholar 

  84. van Beek J, Nicole O, Ali C, Ischenko A, MacKenzie ET, Buisson A, et al. Complement anaphylatoxin C3a is selectively protective against NMDA-induced neuronal cell death. NeuroReport. 2001;12:2289–93. https://doi.org/10.1097/00001756-200102120-00022.

    Article  Google Scholar 

  85. Osaka H, Mukherjee P, Aisen PS, Pasinetti GM. Complement-derived anaphylatoxin C5a protects against glutamate‐mediated neurotoxicity. J Cell Biochem. 1999;73:3303–11.

    Article  Google Scholar 

  86. Shinjyo N, Stahlberg A, Dragunow M, Pekny M, Pekna M. Complement-derived Anaphylatoxin C3a regulates in Vitro differentiation and Migration of neural progenitor cells. Stem Cells. 2009;27(11):2824–32. https://doi.org/10.1002/stem.225.

    Article  CAS  PubMed  Google Scholar 

  87. Mukherjee P, Thomas S, Pasinetti GM. Complement anaphylatoxin C5a neuroprotects through regulation of glutamate receptor subunit 2 in vitro and in vivo. J Neuroinflamm. 2008;5:7. https://doi.org/10.1186/1742-2094-5-5.

    Article  CAS  Google Scholar 

  88. Shutov LP, Warwick CA, Shi X, Gnanasekaran A, Shepherd AJ, Mohapatra DP, et al. The complement system component C5a produces thermal hyperalgesia via macrophage-to-nociceptor signaling that requires NGF and TRPV1. J Neurosci. 2016;36:18.

    Article  Google Scholar 

  89. Chu C, Artis D, Chiu IM. Neuro-immune interactions in the tissues. Immunity. 2020;52:3464–74. https://doi.org/10.1016/j.immuni.2020.02.017.

    Article  CAS  Google Scholar 

  90. Chiu IM, Von Hehn CA, Woolf CJ. Neurogenic inflammation and the peripheral nervous system in host defense and immunopathology. Nat Neurosci. 2012;15:8:1063–7. https://doi.org/10.1038/nn.3144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. DeLeo JA, Tanga FY, Tawfik VL. Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. Neuroscientist. 2004;10:1:40–52. https://doi.org/10.1177/1073858403259950.

    Article  CAS  PubMed  Google Scholar 

  92. Eder A, Dobrowolska, Kamhieh-Milz W. The role of adipose tissue in the Pathogenesis and therapeutic outcomes of inflammatory bowel disease. Cells. 2019;8:6628. https://doi.org/10.3390/cells8060628.

    Article  CAS  Google Scholar 

  93. Ordovas-Montanes J, Rakoff-Nahoum S, Huang S, Riol-Blanco L, Barreiro O, Von Andrian UH. The regulation of immunological processes by peripheral neurons in Homeostasis and Disease. Trends Immunol. 2015;36:10:578–604. https://doi.org/10.1016/j.it.2015.08.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Spallanzani RG, Zemmour D, Xiao TL, Jayewickreme T, Li CR, Bryce PJ, et al. Distinct immunocyte-promoting and adipocyte-generating stromal components coordinate adipose tissue immune and metabolic tenors. Sci Immunol. 2019;4:3513. https://doi.org/10.1126/sciimmunol.aaw3658.

    Article  CAS  Google Scholar 

  95. Mrozek S, Constantin JM, Geeraerts T. Brain-lung crosstalk: implications for neurocritical care patients. World J Crit Care Med. 2015;4:3163–78. https://doi.org/10.5492/wjccm.v4.i3.163.

    Article  Google Scholar 

  96. Chavan SS, Pavlov VA, Tracey KJ. Mechanisms and therapeutic relevance of neuro-immune communication. Immunity. 2017;46:6. https://doi.org/10.1016/j.immuni.2017.06.008.

    Article  CAS  Google Scholar 

  97. Hodo TW, de Aquino MTP, Shimamoto A, Shanker A. Critical neurotransmitters in the Neuroimmune Network. Front Immunol. 2020;11:26. https://doi.org/10.3389/fimmu.2020.01869.

    Article  CAS  Google Scholar 

  98. Prud’homme GJ, Glinka Y, Wang QH. Immunological GABAergic interactions and therapeutic applications in autoimmune diseases. Autoimmun Rev. 2015;14:11:1048–56. https://doi.org/10.1016/j.autrev.2015.07.011.

    Article  CAS  PubMed  Google Scholar 

  99. Liu B, Wang K, Gao HM, Mandavilli B, Wang JY, Hong JS. Molecular consequences of activated microglia in the brain: overactivation induces apoptosis. J Neurochem. 2001;77:1:182–9. https://doi.org/10.1046/j.1471-4159.2001.t01-1-00216.x.

    Article  CAS  PubMed  Google Scholar 

  100. Gaspar R, Soares-Cunha C, Domingues AV, Coimbra B, Baptista FI, Pinto L, et al. Resilience to stress and sex-specific remodeling of microglia and neuronal morphology in a rat model of anxiety and anhedonia. Neurobiol Stress. 2021;14:100302. https://doi.org/10.1016/j.ynstr.2021.100302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Mousa A, Bakhiet M. Role of Cytokine Signaling during Nervous System Development. Int J Mol Sci. 2013;14:7:13931–57. https://doi.org/10.3390/ijms140713931.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Bragazzi NL, Kolahi AA, Nejadghaderi SA, Lochner P, Brigo F, Naldi A, et al. Global, regional, and national burden of Guillain-Barré syndrome and its underlying causes from 1990 to 2019. J Neuroinflammation. 2021;18:1264. https://doi.org/10.1186/s12974-021-02319-4.

    Article  Google Scholar 

  103. Kurman RJ, Shih Ie M. The Dualistic Model of ovarian carcinogenesis: revisited, revised, and expanded. Am J Pathol. 2016;186:4733–47. https://doi.org/10.1016/j.ajpath.2015.11.011.

    Article  Google Scholar 

  104. Feistritzer C, Clausen J, Sturn DH, Djanani A, Gunsilius E, Wiedermann CJ, et al. Natural killer cell functions mediated by the neuropeptide substance P. Regul Pept. 2003;116:1–3. https://doi.org/10.1016/s0167-0115(03)00193-9.

    Article  Google Scholar 

  105. Sabogal-Guáqueta AM, Marmolejo-Garza A, Trombetta-Lima M, Oun A, Hunneman J, Chen T, et al. Species-specific metabolic reprogramming in human and mouse microglia during inflammatory pathway induction. Nat Commun. 2023;14:16454. https://doi.org/10.1038/s41467-023-42096-7.

    Article  CAS  Google Scholar 

  106. Salzer I, Ray S, Schicker K, Boehm S. Nociceptor signalling through ion Channel Regulation via GPCRs. Int J Mol Sci. 2019;20:10. https://doi.org/10.3390/ijms20102488.

    Article  CAS  Google Scholar 

  107. Burnstock G, Boeynaems JM. Purinergic signalling and immune cells. Purinergic Signal. 2014;10:4. https://doi.org/10.1007/s11302-014-9427-2.

    Article  CAS  Google Scholar 

  108. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:6:752–8. https://doi.org/10.1038/nn1472.

    Article  CAS  PubMed  Google Scholar 

  109. Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10:11:1361–8. https://doi.org/10.1038/nn1992.

    Article  CAS  PubMed  Google Scholar 

  110. Zarrinmayeh H, Territo PR. Purinergic receptors of the Central Nervous System: Biology, PET ligands, and their applications. Mol Imaging. 2020;19:26. https://doi.org/10.1177/1536012120927609.

    Article  CAS  Google Scholar 

  111. Donnelly CR, Chen O, Ji RR. How do sensory neurons sense Danger signals? Trends Neurosci. 2020;43. https://doi.org/10.1016/j.tins.2020.07.008. :10:822 – 38.

  112. Vijay K. Toll-like receptors in immunity and inflammatory diseases: past, present, and future. Int Immunopharmacol. 2018;59:391–412. https://doi.org/10.1016/j.intimp.2018.03.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Barajon I, Serrao G, Arnaboldi F, Opizzi E, Ripamonti G, Balsari A, et al. Toll-like receptors 3, 4, and 7 are expressed in the enteric nervous system and dorsal Root Ganglia. J Histochem Cytochemistry. 2009;57(11):1013–23. https://doi.org/10.1369/jhc.2009.953539.

    Article  CAS  Google Scholar 

  114. Yirmiya R, Goshen I. Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun. 2011;25:2:181–213. https://doi.org/10.1016/j.bbi.2010.10.015.

    Article  CAS  PubMed  Google Scholar 

  115. Biesmans S, Meert TF, Bouwknecht JA, Acton PD, Davoodi N, De Haes P, et al. Systemic Immune activation leads to Neuroinflammation and Sickness Behavior in mice. Mediat Inflamm. 2013;2013:14. https://doi.org/10.1155/2013/271359.

    Article  CAS  Google Scholar 

  116. Cornell J, Salinas S, Huang HY, Zhou MO. Microglia regulation of synaptic plasticity and learning and memory. Neural Regen Res. 2022;17:4. https://doi.org/10.4103/1673-5374.322423.

    Article  CAS  Google Scholar 

  117. Delpech JC, Saucisse N, Parkes SL, Lacabanne C, Aubert A, Casenave F, et al. Microglial activation enhances associative taste memory through Purinergic Modulation of glutamatergic neurotransmission. J Neurosci. 2015;35:7:3022–33. https://doi.org/10.1523/jneurosci.3028-14.2015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Bie B, Wu J, Foss JF, Naguib M. Activation of mGluR1 mediates C1q-Dependent Microglial phagocytosis of glutamatergic synapses in Alzheimer’s Rodent models. Mol Neurobiol. 2019;56:8:5568–85. https://doi.org/10.1007/s12035-019-1467-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Afridi R, Suk K. Neuroinflammatory basis of Depression: learning from experimental models. Front Cell Neurosci. 2021;15:9. https://doi.org/10.3389/fncel.2021.691067.

    Article  CAS  Google Scholar 

  120. Zhang LJ, Zhang JQ, You ZL. Switching of the Microglial activation phenotype is a possible treatment for Depression Disorder. Front Cell Neurosci. 2018;12:13. https://doi.org/10.3389/fncel.2018.00306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wohleb ES. Neuron-Microglia Interactions in Mental Health Disorders: for better, and for worse. Front Immunol. 2016;7:13. https://doi.org/10.3389/fimmu.2016.00544.

    Article  CAS  Google Scholar 

  122. Wang HX, He Y, Sun ZL, Ren SY, Liu MX, Wang G, et al. Microglia in depression: an overview of microglia in the pathogenesis and treatment of depression. J Neuroinflamm. 2022;19:126. https://doi.org/10.1186/s12974-022-02492-0.

    Article  Google Scholar 

  123. Yirmiya R, Rimmerman N, Reshef R. Depression as a Microglial Disease. Trends Neurosci. 2015;38:10637–58. https://doi.org/10.1016/j.tins.2015.08.001.

    Article  CAS  Google Scholar 

  124. D’Mello C, Ronaghan N, Zaheer R, Dicay M, Le T, MacNaughton WK, et al. Probiotics improve inflammation-Associated Sickness Behavior by Altering Communication between the Peripheral Immune System and the brain. J Neurosci. 2015;35:30:10821–30. https://doi.org/10.1523/jneurosci.0575-15.2015.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Burton MD, Sparkman NL, Johnson RW. Inhibition of interleukin-6 trans-signaling in the brain facilitates recovery from lipopolysaccharide-induced sickness behavior. J Neuroinflamm. 2011;8:13. https://doi.org/10.1186/1742-2094-8-54.

    Article  CAS  Google Scholar 

  126. Domercq M, Vazquez-Villoldo N, Matute C. Neurotransmitter signaling in the pathophysiology of microglia. Front Cell Neurosci. 2013;7:17. https://doi.org/10.3389/fncel.2013.00049.

    Article  CAS  Google Scholar 

  127. Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR. Microglia in Pain: detrimental and protective roles in Pathogenesis and Resolution of Pain. Neuron. 2018;100:6. https://doi.org/10.1016/j.neuron.2018.11.009.

    Article  CAS  Google Scholar 

  128. Zhuo M, Wu GX, Wu LJ. Neuronal and microglial mechanisms of neuropathic pain. Mol Brain. 2011;4:12. https://doi.org/10.1186/1756-6606-4-31.

    Article  CAS  Google Scholar 

  129. Ward H, West SJ. Microglia: sculptors of neuropathic pain? R Soc Open Sci. 2020;7:623. https://doi.org/10.1098/rsos.200260.

    Article  Google Scholar 

  130. Kohno K, Tsuda M. Role of microglia and P2X4 receptors in chronic pain. Pain Rep. 2021;6:111. https://doi.org/10.1097/pr9.0000000000000864.

    Article  Google Scholar 

  131. Simon DW, McGeachy MJ, Bayır H, Clark RS, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Reviews Neurol. 2017;13:3171–91.

    Google Scholar 

  132. Mazzitelli JA, Smyth LCD, Cross KA, Dykstra T, Sun J, Du S, et al. Cerebrospinal fluid regulates skull bone marrow niches via direct access through dural channels. Nat Neurosci. 2022;25:5555–60. https://doi.org/10.1038/s41593-022-01029-1.

    Article  CAS  Google Scholar 

  133. Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, et al. Single-cell RNA sequencing of Microglia throughout the mouse lifespan and in the injured brain reveals Complex Cell-State changes. Immunity. 2019;50:1253–e716. https://doi.org/10.1016/j.immuni.2018.11.004.

    Article  CAS  Google Scholar 

  134. Eyo UB, Peng JY, Swiatkowski P, Mukherjee A, Bispo A, Wu LJ. Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after Status Epilepticus. J Neurosci. 2014;34:32:10528–40. https://doi.org/10.1523/jneurosci.0416-14.2014.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev. 2011;91:2:461–553. https://doi.org/10.1152/physrev.00011.2010.

    Article  CAS  PubMed  Google Scholar 

  136. Farber K, Kettenmann H. Functional role of calcium signals for microglial function. Glia. 2006;54:7656–65. https://doi.org/10.1002/glia.20412.

    Article  Google Scholar 

  137. Luo LX, Song SS, Ezenwukwa CC, Jalali S, Sun BS, Sun DD. Ion channels and transporters in microglial function in physiology and brain diseases. Neurochem Int. 2021;142:12. https://doi.org/10.1016/j.neuint.2020.104925.

    Article  CAS  Google Scholar 

  138. Liu GJ, Nagarajah R, Banati RB, Bennett MR. Glutamate induces directed chemotaxis of microglia. Eur J Neurosci. 2009;29:6:1108–18. https://doi.org/10.1111/j.1460-9568.2009.06659.x.

    Article  PubMed  Google Scholar 

  139. Liu H, Leak RK, Hu X. Neurotransmitter receptors on microglia. BMJ. 2016;1:2. https://doi.org/10.1136/svn-2016-000012.

    Article  Google Scholar 

  140. Pocock JM, Kettenmann H. Neurotransmitter receptors on microglia. Trends Neurosci. 2007;30:10. https://doi.org/10.1016/j.tins.2007.07.007.

    Article  CAS  Google Scholar 

  141. Garaschuk O, Verkhratsky A. Physiology of Microglia. In: Garaschuk O, Verkhratsky A, editors. Microglia: methods and protocols. Methods in Molecular Biology. Totowa: Humana Press Inc; 2019. pp. 27–40. https://doi.org/10.1007/978-1-4939-9658-2_3.

    Chapter  Google Scholar 

  142. York EM, Bernier LP, MacVicar BA. Microglial modulation of neuronal activity in the healthy brain. Dev Neurobiol. 2018;78:6.

    Article  Google Scholar 

  143. Kato G, Inada H, Wake H, Akiyoshi A, Miyamoto R, Eto K, et al. Microglial contact prevents excess depolarization and rescues neurons from Excitotoxicity. Eneuro. 2016;3(3):ENEURO0004–162016. https://doi.org/10.1523/Eneuro.0004-16.2016.

    Article  CAS  Google Scholar 

  144. Taylor DL, Diemel LT, Pocock JM. Activation of microglial group III metabotropic glutamate receptors protects neurons against microglial neurotoxicity. J Neurosci. 2003;23:6.

    Article  Google Scholar 

  145. Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional microglia-neuron communication in Health and Disease. Front Cell Neurosci. 2018;12:26. https://doi.org/10.3389/fncel.2018.00323.

    Article  CAS  Google Scholar 

  146. Vinet J, van Weering HRJ, Heinrich A, Kalin RE, Wegner A, Brouwer N, et al. Neuroprotective function for ramified microglia in hippocampal excitotoxicity. J Neuroinflamm. 2012;9:15. https://doi.org/10.1186/1742-2094-9-27.

    Article  CAS  Google Scholar 

  147. Jantzie LL, Talos DM, Jackson MC, Park HK, Graham DA, Lechpammer M, et al. Developmental expression of N-Methyl-D-Aspartate (NMDA) receptor subunits in Human White and Gray Matter: potential mechanism of increased vulnerability in the immature brain. Cereb Cortex. 2015;25:2482–95. https://doi.org/10.1093/cercor/bht246.

    Article  Google Scholar 

  148. Ellis A, Bennett DLH. Neuroinflammation and the generation of neuropathic pain. Br J Anaesth. 2013;111:1:26–37. https://doi.org/10.1093/bja/aet128.

    Article  CAS  PubMed  Google Scholar 

  149. Assas BM, Pennock JI, Miyan JA. Calcitonin gene-related peptide is a key neurotransmitter in the neuro-immune axis. Front NeuroSci. 2014;8:23.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Perner C, Flayer CH, Zhu X, Aderhold PA, Dewan ZNA, Voisin T, et al. Substance P release by sensory neurons triggers Dendritic Cell Migration and initiates the Type-2 Immune response to Allergens. Immunity. 2020;53(5):1063–e777. https://doi.org/10.1016/j.immuni.2020.10.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Szabo-Pardi TA, Barron LR, Lenert ME, Burton MD. Sensory neuron TLR4 mediates the development of nerve-injury induced mechanical hypersensitivity in female mice. Brain Behav Immun. 2021;97:42–60. https://doi.org/10.1016/j.bbi.2021.06.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Cowie AM, Menzel AD, Oharaa C, Lawlor MW, Stucky CL. NOD-like receptor protein 3 inflammasome drives postoperative mechanical pain in a sex-dependent manner. Pain. 2019;160:8. https://doi.org/10.1097/j.pain.0000000000001555.

    Article  CAS  Google Scholar 

  153. Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature. 2013;501:746552–7. https://doi.org/10.1038/nature12479.

    Article  CAS  Google Scholar 

  154. Wang T, Ma C. Peripheral nociceptors as Immune sensors in the Development of Pain and Itch. Advances in Experimental Medicine and Biology. Springer Netherlands; 2016. pp. 77–85. https://doi.org/10.1007/978-94-017-7537-3_6.

  155. Liu Y, Caterina MJ, Qu LT. Sensory Neuron expressed fc gamma RI mediates Postinflammatory Arthritis Pain in Female mice. Front Immunol. 2022;13:14. https://doi.org/10.3389/fimmu.2022.889286.

    Article  CAS  Google Scholar 

  156. Jiang HW, Shen XH, Chen ZY, Liu F, Wang T, Xie YK, et al. Nociceptive neuronal Fc-gamma receptor I is involved in IgG immune complex induced pain in the rat. Brain Behav Immun. 2017;62:351–61. https://doi.org/10.1016/j.bbi.2017.03.001.

    Article  CAS  PubMed  Google Scholar 

  157. Wang L, Jiang XH, Zheng Q, Jeon SM, Chen T, Liu Y, et al. Neuronal fc gamma RI mediates acute and chronic joint pain. J Clin Invest. 2019;129:9:3754–69. https://doi.org/10.1172/jci128010.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Goebel A, Krock E, Gentry C, Israel MR, Jurczak A, Urbina CM, et al. Passive transfer of fibromyalgia symptoms from patients to mice. J Clin Invest. 2021;131:13. https://doi.org/10.1172/jci144201.

    Article  CAS  Google Scholar 

  159. Qu L, Zhang P, Lamotte RH, Ma C, Brain. Behav Immun. 2011;25:7. https://doi.org/10.1016/j.bbi.2011.04.008.

    Article  CAS  Google Scholar 

  160. Bersellini Farinotti A, Wigerblad G, Nascimento D, Bas DB, Morado Urbina C, Nandakumar KS, et al. Cartilage-binding antibodies induce pain through immune complex–mediated activation of neurons. J Exp Med. 2019;216:8. https://doi.org/10.1084/jem.20181657.

    Article  Google Scholar 

  161. Corr M, Crain B. The role of fc gamma R signaling in the K/B x N serum transfer model of arthritis. J Immunol. 2002;169:11:6604–9. https://doi.org/10.4049/jimmunol.169.11.6604.

    Article  CAS  PubMed  Google Scholar 

  162. Osborne BF, Turano A, Schwarz JM. Sex differences in the neuroimmune system. Curr Opin Behav Sci. 2018;23:118–23. https://doi.org/10.1016/j.cobeha.2018.05.007.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Gregus AM, Levine IS, Eddinger KA, Yaksh TL, Buczynski MW. Sex differences in neuroimmune and glial mechanisms of pain. Pain. 2021;162:8. https://doi.org/10.1097/j.pain.0000000000002215.

    Article  CAS  Google Scholar 

  164. Cowie AM, Dittel BN, Stucky CL. A novel sex-dependent target for the treatment of Postoperative Pain: the NLRP3 inflammasome. Front Neurol. 2019;10:9. https://doi.org/10.3389/fneur.2019.00622.

    Article  Google Scholar 

  165. Scheff NN, Bhattacharya A, Dowse E, Dang RX, Dolan JC, Wang S, et al. Neutrophil-mediated endogenous Analgesia contributes to sex differences in oral Cancer Pain. Front Integr Neurosci. 2018;12:15. https://doi.org/10.3389/fnint.2018.00052.

    Article  CAS  Google Scholar 

  166. Sorge RE, Mapplebeck JCS, Rosen S, Beggs S, Taves S, Alexander JK, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18:8. https://doi.org/10.1038/nn.4053.

    Article  CAS  Google Scholar 

  167. Inyang KE, Szabo-Pardi T, Wentworth E, Mcdougal TA, Dussor G, Burton MD, et al. The antidiabetic drug metformin prevents and reverses neuropathic pain and spinal cord microglial activation in male but not female mice. Pharmacol Res. 2019;139:1–16. https://doi.org/10.1016/j.phrs.2018.10.027.

    Article  CAS  PubMed  Google Scholar 

  168. Yang JX, Wang HF, Chen JZ, Li HY, Hu JC, Yu AA, et al. Potential Neuroimmune Interaction in Chronic Pain: a review on Immune cells in Peripheral and Central Sensitization. Front Pain Res (Lausanne). 2022;3:946846. https://doi.org/10.3389/fpain.2022.946846.

    Article  PubMed  Google Scholar 

  169. Laumet G, Edralin JD, Dantzer R, Heijnen CJ, Kavelaars A. CD3(+) T cells are critical for the resolution of comorbid inflammatory pain and depression-like behavior. Neurobiol Pain. 2020;7:100043. https://doi.org/10.1016/j.ynpai.2020.100043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Lenert ME, Szabo-Pardi TA, Burton MD. Regulatory T-cells and IL-5 mediate pain outcomes in a preclinical model of chronic muscle pain. Mol Pain. 2023;19:17448069221110691. https://doi.org/10.1177/17448069221110691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Rosen SF, Ham B, Haichin M, Walters IC, Tohyama S, Sotocinal SG, et al. Increased pain sensitivity and decreased opioid analgesia in T-cell-deficient mice and implications for sex differences. Pain. 2019;160:2358–66. https://doi.org/10.1097/j.pain.0000000000001420.

    Article  CAS  Google Scholar 

  172. Mifflin KA, Yousuf MS, Thorburn KC, Huang J, Perez-Munoz ME, Tenorio G, et al. Voluntary wheel running reveals sex-specific nociceptive factors in murine experimental autoimmune encephalomyelitis. Pain. 2019;160:4870–81. https://doi.org/10.1097/j.pain.0000000000001465.

    Article  CAS  Google Scholar 

  173. Chernov AV, Hullugundi SK, Eddinger KA, Dolkas J, Remacle AG, Angert M, et al. A myelin basic protein fragment induces sexually dimorphic transcriptome signatures of neuropathic pain in mice. J Biol Chem. 2020;295:31:10807–21. https://doi.org/10.1074/jbc.RA120.013696.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Kuhn JA, Vainchtein ID, Braz J, Hamel K, Bernstein M, Craik V, et al. Regulatory T-cells inhibit microglia-induced pain hypersensitivity in female mice. eLife. 2021;10. https://doi.org/10.7554/elife.69056.

  175. Rosen SF, Ham B, Drouin S, Boachie N, Chabot-Dore AJ, Austin JS, et al. T-Cell mediation of pregnancy Analgesia Affecting Chronic Pain in mice. J Neurosci. 2017;37:41:9819–27. https://doi.org/10.1523/jneurosci.2053-17.2017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Lopes DM, Malek N, Edye M, Jager SB, McMurray S, McMahon SB, et al. Sex differences in peripheral not central immune responses to pain-inducing injury. Sci Rep. 2017;7:116460. https://doi.org/10.1038/s41598-017-16664-z.

    Article  CAS  Google Scholar 

  177. Guan Z, Kuhn JA, Wang X, Colquitt B, Solorzano C, Vaman S, et al. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat Neurosci. 2016;19:1:94–101. https://doi.org/10.1038/nn.4189.

    Article  CAS  PubMed  Google Scholar 

  178. Korczeniewska OA, Husain S, Khan J, Eliav E, Soteropoulos P, Benoliel R. Differential gene expression in trigeminal ganglia of male and female rats following chronic constriction of the infraorbital nerve. Eur J Pain. 2018;22:5875–88. https://doi.org/10.1002/ejp.1174.

    Article  CAS  Google Scholar 

  179. Kuhn JA, Vainchtein ID, Braz J, Hamel K, Bernstein M, Craik V, et al. Regulatory T-cells inhibit microglia-induced pain hypersensitivity in female mice. Elife. 2021;10:17. https://doi.org/10.7554/eLife.69056.sa1.

    Article  Google Scholar 

  180. Chen G, Luo X, Qadri MY, Berta T, Ji R-R. Sex-dependent Glial Signaling in Pathological Pain: distinct roles of spinal microglia and astrocytes. Neurosci Bull. 2018;34:1:98–108. https://doi.org/10.1007/s12264-017-0145-y.

    Article  CAS  PubMed  Google Scholar 

  181. Mapplebeck JCS, Beggs S, Salter MW. Sex differences in pain: a tale of two immune cells. Pain. 2016;157(2):S2–6. https://doi.org/10.1097/j.pain.0000000000000389.

    Article  PubMed  Google Scholar 

  182. Yu XB, Liu HJ, Hamel KA, Morvan MG, Yu S, Leff J, et al. Dorsal root ganglion macrophages contribute to both the initiation and persistence of neuropathic pain. Nat Commun. 2020;11:112. https://doi.org/10.1038/s41467-019-13839-2.

    Article  CAS  Google Scholar 

  183. Sorge RE, Lacroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin J-S, Ritchie J, et al. Spinal cord toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci. 2011;31:43:15450–4. https://doi.org/10.1523/jneurosci.3859-11.2011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Gay NJ, Keith FJ. Drosophila toll and IL-1 receptor. Nature. 1991;351:6325:355–6. https://doi.org/10.1038/351355b0.

    Article  CAS  PubMed  Google Scholar 

  185. Uematsu S, Akira S. Toll-like receptors (TLRs) and their ligands. Handb Exp Pharmacol. 2008;183:1–20. https://doi.org/10.1007/978-3-540-72167-3_1.

    Article  CAS  Google Scholar 

  186. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:7:499–511. https://doi.org/10.1038/nri1391.

    Article  CAS  PubMed  Google Scholar 

  187. Taguchi T, Mitcham JL, Dower SK, Sims JE, Testa JR. Chromosomal localization of TIL, a gene encoding a protein related to the Drosophila transmembrane receptor toll, to human chromosome 4p14. Genomics. 1996;32(3):486–8. https://doi.org/10.1006/geno.1996.0150.

    Article  CAS  PubMed  Google Scholar 

  188. Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the Drosophila toll protein signals activation of adaptive immunity. Nature. 1997;388:6640:394–7. https://doi.org/10.1038/41131.

    Article  CAS  PubMed  Google Scholar 

  189. Raetz CR, Whitfield C. Lipopolysaccharide endotoxins. Annu Rev Biochem. 2002;71:635–700. https://doi.org/10.1146/annurev.biochem.71.110601.135414.

    Article  CAS  PubMed  Google Scholar 

  190. Huang S, Rutkowsky JM, Snodgrass RG, Ono-Moore KD, Schneider DA, Newman JW, et al. Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J Lipid Res. 2012;53:9. https://doi.org/10.1194/jlr.D029546.

    Article  CAS  Google Scholar 

  191. Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff HH. Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis. 2016;244:211–5. https://doi.org/10.1016/j.atherosclerosis.2015.11.015.

    Article  CAS  PubMed  Google Scholar 

  192. Olson JK, Miller SD. Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs. J Immunol. 2004;173:6:3916–24. https://doi.org/10.4049/jimmunol.173.6.3916.

    Article  CAS  PubMed  Google Scholar 

  193. Bowman CC, Rasley A, Tranguch SL, Marriott I. Cultured astrocytes express toll-like receptors for bacterial products. Glia. 2003;43:3. https://doi.org/10.1002/glia.10256.

    Article  Google Scholar 

  194. Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, Jo DG, et al. Pivotal role for neuronal toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A. 2007;104:34. https://doi.org/10.1073/pnas.0702553104.

    Article  Google Scholar 

  195. Kawai T, Adachi O, Ogawa T, Takeda K, Akira S. Unresponsiveness of MyD88-deficient mice to endotoxin. Immunity. 1999;11:1115–22. https://doi.org/10.1016/s1074-7613(00)80086-2.

    Article  Google Scholar 

  196. Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, et al. Cutting edge: toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the lps gene product. J Immunol. 1999;162:7.

    Article  Google Scholar 

  197. Lu Z, Li Y, Jin J, Zhang X, Lopes-Virella MF, Huang Y. Toll-like receptor 4 activation in microvascular endothelial cells triggers a robust inflammatory response and cross talk with mononuclear cells via interleukin-6. Arterioscler Thromb Vasc Biol. 2012;32:71696–706. https://doi.org/10.1161/ATVBAHA.112.251181.

    Article  CAS  Google Scholar 

  198. Bhattacharyya S, Wang W, Qin W, Cheng K, Coulup S, Chavez S, et al. TLR4-dependent fibroblast activation drives persistent organ fibrosis in skin and lung. JCI Insight. 2018;3:13. https://doi.org/10.1172/jci.insight.98850.

    Article  Google Scholar 

  199. Min H, Cho WH, Lee H, Choi B, Kim YJ, Lee HK, et al. Association of TRPV1 and TLR4 through the TIR domain potentiates TRPV1 activity by blocking activation-induced desensitization. Mol Pain. 2018;14:1744806918812636. https://doi.org/10.1177/1744806918812636.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Michot B, Casey SM, Lee CS, Erdogan O, Basu H, Chiu I, et al. Lipopolysaccharide-Induced TRPA1 upregulation in trigeminal neurons is dependent on TLR4 and vesicular exocytosis. J Neurosci. 2023;43:40:6731–44. https://doi.org/10.1523/jneurosci.0162-23.2023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Diogenes A, Ferraz C, Akopian Aa, Henry M, Hargreaves K. LPS sensitizes TRPV1 via activation of TLR4 in trigeminal sensory neurons. J Dent Res. 2011;90:6.

    Article  Google Scholar 

  202. Xie MX, Zhang XL, Xu J, Zeng WA, Li D, Xu T, et al. Nuclear factor-kappab gates na(v)1.7 channels in DRG neurons via protein-protein Interaction. iScience. 2019;19:623–33. https://doi.org/10.1016/j.isci.2019.08.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Guo LH, Schluesener HJ. The innate immunity of the central nervous system in chronic pain: the role of toll-like receptors. Cell Mol Life Sci. 2007;64:9:1128–36. https://doi.org/10.1007/s00018-007-6494-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Heiman A, Pallottie A, Heary RF, Elkabes S. Toll-like receptors in central nervous system injury and disease: a focus on the spinal cord. Brain Behav Immun. 2014;42:232–45. https://doi.org/10.1016/j.bbi.2014.06.203.

    Article  CAS  PubMed  Google Scholar 

  205. Raghavendra V, Tanga FY, DeLeo JA. Complete freunds adjuvant-induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci. 2004;20:2. https://doi.org/10.1111/j.1460-9568.2004.03514.x.

    Article  Google Scholar 

  206. Zhao XH, Zhang T, Li YQ. The up-regulation of spinal toll-like receptor 4 in rats with inflammatory pain induced by complete Freund’s adjuvant. Brain Res Bull. 2015;111:97–103. https://doi.org/10.1016/j.brainresbull.2015.01.002.

    Article  CAS  PubMed  Google Scholar 

  207. Agalave NM, Rudjito R, Farinotti AB, Khoonsari PE, Sandor K, Nomura Y, et al. Sex-dependent role of microglia in disulfide high mobility group box 1 protein-mediated mechanical hypersensitivity. Pain. 2021;162:2446–58. https://doi.org/10.1097/j.pain.0000000000002033.

    Article  CAS  Google Scholar 

  208. Woller SA, Ocheltree C, Wong SY, Bui A, Fujita Y, dos Santos GG, et al. Neuraxial TNF and IFN-beta co-modulate persistent allodynia in arthritic mice. Brain Behav Immun. 2019;76:151–8. https://doi.org/10.1016/j.bbi.2018.11.014.

    Article  CAS  PubMed  Google Scholar 

  209. Christianson CA, Dumlao DS, Stokes JA, Dennis EA, Svensson CI, Corr M, et al. Spinal TLR4 mediates the transition to a persistent mechanical hypersensitivity after the resolution of inflammation in serum-transferred arthritis. Pain. 2011;152:12:2881–91. https://doi.org/10.1016/j.pain.2011.09.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Zaks-Zilberman M, Zaks TZ, Vogel SN. Induction of proinflammatory and chemokine genes by lipopolysaccharide and paclitaxel (taxol) in murine and human breast cancer cell lines. Cytokine. 2001;15:3156–65. https://doi.org/10.1006/cyto.2001.0935.

    Article  CAS  Google Scholar 

  211. O’Brien JM, Wewers MD, Moore SA, Allen JN. Taxol and colchicine increase LPS-induced pro-IL-1 beta production, but do not increase IL-1 beta secretion. A role for microtubules in the regulation of IL-1 beta production. J Immunol. 1995;154:8. https://www.ncbi.nlm.nih.gov/pubmed/7706748.

    Google Scholar 

  212. Li Y, Zhang H, Kosturakis AK, Jawad AB, Dougherty PM. Toll-like receptor 4 signaling contributes to Paclitaxel-induced peripheral neuropathy. J Pain. 2014;15:7. https://doi.org/10.1016/j.jpain.2014.04.001.

    Article  CAS  Google Scholar 

  213. Illias AM, Yu KJ, Hwang SH, Solis J, Zhang H, Velasquez JF, et al. Dorsal root ganglion toll-like receptor 4 signaling contributes to oxaliplatin-induced peripheral neuropathy. Pain. 2022;163:5923–35. https://doi.org/10.1097/j.pain.0000000000002454.

    Article  CAS  Google Scholar 

  214. Araldi D, Bogen O, Green PG, Levine JD. Role of nociceptor toll-like receptor 4 (TLR4) in opioid-induced hyperalgesia and hyperalgesic priming. J Neurosci. 2019;39:33.

    Article  Google Scholar 

  215. Mercadante S, Arcuri E, Santoni A. Opioid-induced tolerance and hyperalgesia. CNS Drugs. 2019;33:943–55. https://doi.org/10.1007/s40263-019-00660-0.

    Article  CAS  PubMed  Google Scholar 

  216. Weber L, Yeomans DC, Tzabazis A. Opioid-induced hyperalgesia in clinical anesthesia practice: what has remained from theoretical concepts and experimental studies? Curr Opin Anaesthesiol. 2017;30:4458–65. https://doi.org/10.1097/ACO.0000000000000485.

    Article  CAS  Google Scholar 

  217. Li J, Csakai A, Jin J, Zhang F, Yin H. Therapeutic developments targeting toll-like receptor-4-Mediated Neuroinflammation. ChemMedChem. 2016;11:2. https://doi.org/10.1002/cmdc.201500188.

    Article  CAS  Google Scholar 

  218. Ellis A, Grace PM, Wieseler J, Favret J, Springer K, Skarda B, et al. Morphine amplifies mechanical allodynia via TLR4 in a rat model of spinal cord injury. Brain Behav Immun. 2016;58:348–56. https://doi.org/10.1016/j.bbi.2016.08.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Tanga FY, Nutile-McMenemy N, DeLeo JA. The CNS role of toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci U S A. 2005;102:16:5856–61. https://doi.org/10.1073/pnas.0501634102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Hutchinson MR, Zhang Y, Brown K, Coats BD, Shridhar M, Sholar PW, et al. Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: involvement of toll-like receptor 4 (TLR4). Eur J Neurosci. 2008;28:120–9. https://doi.org/10.1111/j.1460-9568.2008.06321.x.

    Article  Google Scholar 

  221. Bettoni I, Comelli F, Rossini C, Granucci F, Giagnoni G, Peri F, et al. Glial TLR4 receptor as new target to treat neuropathic pain: efficacy of a new receptor antagonist in a model of peripheral nerve injury in mice. Glia. 2008;56:12:1312–9. https://doi.org/10.1002/glia.20699.

    Article  PubMed  Google Scholar 

  222. Bruno K, Woller SA, Miller YI, Yaksh TL, Wallace M, Beaton G, et al. Targeting toll-like receptor-4 (TLR4)-an emerging therapeutic target for persistent pain states. Pain. 2018;159:10. https://doi.org/10.1097/j.pain.0000000000001306.

    Article  CAS  Google Scholar 

  223. Woller SA, Ravula SB, Tucci FC, Beaton G, Corr M, Isseroff RR, et al. 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 Behav Immun. 2016;56:271–80. https://doi.org/10.1016/j.bbi.2016.03.026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. 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. J Neuroinflammation. 2013;10:148. https://doi.org/10.1186/1742-2094-10-148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Ghosh C, Bishayi B. Characterization of toll-like Receptor-4 (TLR-4) in the Spleen and Thymus of Swiss albino mice and its modulation in experimental endotoxemia. J Immunol Res. 2015;2015:13. https://doi.org/10.1155/2015/137981.

    Article  CAS  Google Scholar 

  226. Mogil JS, Wilson SG, Bon K, Lee SE, Chung K, Raber P, et al. Heritability of nociception I: responses of 11 inbred mouse strains on 12 measures of nociception. Pain. 1999;80(1–2):67–82. https://doi.org/10.1016/s0304-3959(98)00197-3.

    Article  CAS  PubMed  Google Scholar 

  227. Qureshi ST, Larivière L, Leveque G, Clermont S, Moore KJ, Gros P, et al. Endotoxin-tolerant mice have mutations in toll-like receptor 4 (< i > Tlr4). J Exp Med. 1999;189:4615–25. https://doi.org/10.1084/jem.189.4.615.

    Article  Google Scholar 

  228. Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, et al. Cutting edge:: toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide:: evidence for TLR4 as the < i > lps gene product. J Immunol. 1999;162:7. ://WOS:000079278000001.

    Article  Google Scholar 

  229. Nedrebo T, Reed RK. Different serotypes of endotoxin (lipopolysaccharide) cause different increases in albumin extravasation in rats. Shock. 2002;18:2. https://doi.org/10.1097/00024382-200208000-00008.

    Article  Google Scholar 

  230. Akarsu ES, Mamuk S. Escherichia coli lipopolysaccharides produce serotype-specific hypothermic response in biotelemetered rats. Am J Physiol-Regul Integr Comp Physiol. 2007;292(5):R1846–50. https://doi.org/10.1152/ajpregu.00786.2006.

    Article  CAS  PubMed  Google Scholar 

  231. Migale R, Herbert BR, Lee YS, Sykes L, Waddington SN, Peebles D, et al. Specific lipopolysaccharide serotypes induce Differential maternal and neonatal inflammatory responses in a murine model of Preterm Labor. Am J Pathol. 2015;185:92390–401. https://doi.org/10.1016/j.ajpath.2015.05.015.

    Article  CAS  Google Scholar 

  232. Huck NA, Siliezar-Doyle J, Haight ES, Ishida R, Forman TE, Wu S, et al. Temporal contribution of myeloid-lineage TLR4 to the transition to Chronic Pain: a focus on sex differences. J Neurosci. 2021;41:19:4349–65. https://doi.org/10.1523/JNEUROSCI.1940-20.2021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Alexander SN, Reed OA, Burton MD. Spinal cord microglia drive sex differences in ethanol-mediated PGE2-induced allodynia. Brain Behav Immun. 2024;122:399–421. https://doi.org/10.1016/j.bbi.2024.08.026.

    Article  CAS  PubMed  Google Scholar 

  234. Kwak MS, Kim HS, Lee B, Kim YH, Son M, Shin JS. Immunological significance of HMGB1 post-translational modification and Redox Biology. Front Immunol. 2020;11:1189. https://doi.org/10.3389/fimmu.2020.01189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem. 1995;270:43:25752–61. https://doi.org/10.1074/jbc.270.43.25752.

    Article  CAS  PubMed  Google Scholar 

  236. Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, et al. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 2004;279:9:7370–7. https://doi.org/10.1074/jbc.M306793200.

    Article  CAS  PubMed  Google Scholar 

  237. Thorburn J, Horita H, Redzic J, Hansen K, Frankel AE, Thorburn A. Autophagy regulates selective HMGB1 release in tumor cells that are destined to die. Cell Death Differ. 2009;16:1175–83. https://doi.org/10.1038/cdd.2008.143.

    Article  Google Scholar 

  238. Qin S, Wang H, Yuan R, Li H, Ochani M, Ochani K, et al. Role of HMGB1 in apoptosis-mediated sepsis lethality. J Exp Med. 2006;203:7:1637–42. https://doi.org/10.1084/jem.20052203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:6894:191–5. https://doi.org/10.1038/nature00858.

    Article  CAS  PubMed  Google Scholar 

  240. Venereau E, Schiraldi M, Uguccioni M, Bianchi ME. HMGB1 and leukocyte migration during trauma and sterile inflammation. Mol Immunol. 2013;55:1:76–82. https://doi.org/10.1016/j.molimm.2012.10.037.

    Article  CAS  PubMed  Google Scholar 

  241. Yamasoba D, Tsubota M, Domoto R, Sekiguchi F, Nishikawa H, Liu K, et al. Peripheral HMGB1-induced hyperalgesia in mice: Redox state-dependent distinct roles of RAGE and TLR4. J Pharmacol Sci. 2016;130:2139–42. https://doi.org/10.1016/j.jphs.2016.01.005.

    Article  CAS  Google Scholar 

  242. Müller S, Ronfani L, Bianchi ME. Regulated expression and subcellular localization of HMGB1, a chromatin protein with a cytokine function. J Intern Med. 2004;255. https://doi.org/10.1111/j.1365-2796.2003.01296.x. :3:332 – 43.

  243. Shibasaki M, Sasaki M, Miura M, Mizukoshi K, Ueno H, Hashimoto S, et al. Induction of high mobility group box-1 in dorsal root ganglion contributes to pain hypersensitivity after peripheral nerve injury. Pain. 2010;149:3514–21. https://doi.org/10.1016/j.pain.2010.03.023.

    Article  CAS  Google Scholar 

  244. Feldman P, Due MR, Ripsch MS, Khanna R, White FA. The persistent release of HMGB1 contributes to tactile hyperalgesia in a rodent model of neuropathic pain. J Neuroinflammation. 2012;9:180. https://doi.org/10.1186/1742-2094-9-180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Agalave NM, Larsson M, Abdelmoaty S, Su J, Baharpoor A, Lundback P, et al. Spinal HMGB1 induces TLR4-mediated long-lasting hypersensitivity and glial activation and regulates pain-like behavior in experimental arthritis. Pain. 2014;155:9:1802–13. https://doi.org/10.1016/j.pain.2014.06.007.

    Article  CAS  PubMed  Google Scholar 

  246. Rudjito R, Agalave NM, Farinotti AB, Lundback P, Szabo-Pardi TA, Price TJ, et al. Sex- and cell-dependent contribution of peripheral high mobility group box 1 and TLR4 in arthritis-induced pain. Pain. 2021;162:2459–70. https://doi.org/10.1097/j.pain.0000000000002034.

    Article  CAS  Google Scholar 

  247. Yang H, Zeng Q, Silverman HA, Gunasekaran M, George SJ, Devarajan A, et al. HMGB1 released from nociceptors mediates inflammation. Proc Natl Acad Sci U S A. 2021;118:33. https://doi.org/10.1073/pnas.2102034118.

    Article  CAS  Google Scholar 

  248. Lenert ME, Avona A, Garner KM, Barron LR, Burton MD. Sensory neurons, Neuroimmunity, and Pain Modulation by Sex hormones. Endocrinology. 2021;162:817. https://doi.org/10.1210/endocr/bqab109.

    Article  CAS  Google Scholar 

  249. Payrits M, Sághy É, Csekő K, Pohóczky K, Bölcskei K, Ernszt D, et al. Estradiol sensitizes the Transient Receptor Potential Vanilloid 1 receptor in Pain responses. Endocrinology. 2017;158:10:3249–58. https://doi.org/10.1210/en.2017-00101.

    Article  CAS  PubMed  Google Scholar 

  250. Seol SH, Chung G. Review article Estrogen-dependent regulation of transient receptor potential vanilloid 1 (TRPV1) and P2X purinoceptor 3 (P2X3): Implication in burning mouth syndrome. J Dent Sci. 2022;17(1):8–13. https://doi.org/10.1016/j.jds.2021.06.007.

    Article  PubMed  Google Scholar 

  251. Cho T, Chaban VV. Expression of P2X3 and TRPV1 receptors in primary sensory neurons from estrogen receptors-alpha and estrogen receptor-beta knockout mice. NeuroReport. 2012;23:9:530–4. https://doi.org/10.1097/WNR.0b013e328353fabc.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Chen SC, Chang TJ, Wu FS. Competitive inhibition of the capsaicin receptor-mediated current by dehydroepiandrosterone in rat dorsal root ganglion neurons. J Pharmacol Exp Ther. 2004;311:2. https://doi.org/10.1124/jpet.104.069096.

    Article  CAS  Google Scholar 

  253. Mendez-Resendiz KA, Enciso-Pablo O, Gonzalez-Ramirez R, Juarez-Contreras R, Rosenbaum T, Morales-Lazaro SL. Steroids and TRP channels: a close relationship. Int J Mol Sci. 2020;21:1136. https://doi.org/10.3390/ijms21113819.

    Article  CAS  Google Scholar 

  254. Apkhazava M, Kvachadze I, Tsagareli M, Mzhavanadze D, Chakhnashvili M. The Relationship between Thermal Pain Sensation, Free Testosterone, TRPV1, MOR levels and various degrees of hostility in Young Healthy males. Georgian Med News. 2018;283(109–14). ://MEDLINE:30516504.

  255. Desai MK, Brinton RD. Autoimmune disease in women: endocrine transition and risk across the Lifespan. Front Endocrinol. 2019;10:19. https://doi.org/10.3389/fendo.2019.00265.

    Article  Google Scholar 

  256. Liva SM, Voskuhl RR. Testosterone acts directly on CD4(+) T lymphocytes to increase IL-10 production. J Immunol. 2001;167:4. https://doi.org/10.4049/jimmunol.167.4.2060.

    Article  Google Scholar 

  257. Angum F, Khan T, Kaler J, Siddiqui L, Hussain A. The prevalence of Autoimmune disorders in women: a narrative review. Cureus. 2020. https://doi.org/10.7759/cureus.8094.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Bartley EJ, Fillingim RB. Sex differences in pain: a brief review of clinical and experimental findings. Br J Anaesth. 2013;111:1. https://doi.org/10.1093/bja/aet127.

    Article  Google Scholar 

  259. Lesnak JB, Inoue S, Lima L, Rasmussen L, Sluka KA. Testosterone protects against the development of widespread muscle pain in mice. Pain. 2020;161:122898–908. https://doi.org/10.1097/j.pain.0000000000001985.

    Article  CAS  Google Scholar 

  260. Sorge RE, Mapplebeck JCS, Rosen S, Beggs S, Taves S, Alexander JK, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18:81081–. https://doi.org/10.1038/nn.4053.

    Article  CAS  Google Scholar 

  261. Rosen S, Ham B, Mogil JS. Sex differences in neuroimmunity and pain. J Neurosci Res. 2017;95(1–2):500–8. https://doi.org/10.1002/jnr.23831.

    Article  CAS  PubMed  Google Scholar 

  262. Edinger KL, Frye CA. Testosterone’s analgesic, anxiolytic, and cognitive-enhancing effects may be due in part to actions of its 5 alpha-reduced metabolites in the hippocampus. Behav Neurosci. 2004;118:6:1352–64. https://doi.org/10.1037/0735-7044.118.6.1352.

    Article  CAS  PubMed  Google Scholar 

  263. Edinger KL, Frye CA. Testosterone’s anti-anxiety and analgesic effects may be due in part to actions of its 5 alpha-reduced metabolites in the hippocampus. Psychoneuroendocrinology. 2005;30:5. https://doi.org/10.1016/j.psyneuen.2004.11.001.

    Article  CAS  Google Scholar 

  264. Lee KS, Zhang Y, Asgar J, Auh Q-S, Chung M-K, Ro JY. Androgen receptor transcriptionally regulates µ-opioid receptor expression in rat trigeminal ganglia. Neuroscience. 2016;331:52–61. https://doi.org/10.1016/j.neuroscience.2016.06.023.

    Article  CAS  PubMed  Google Scholar 

  265. Bai X, Zhang X, Zhou Q. Effect of testosterone on TRPV1 expression in a model of Orofacial Myositis Pain in the rat. J Mol Neurosci. 2018;64:1:93–101. https://doi.org/10.1007/s12031-017-1009-7.

    Article  CAS  PubMed  Google Scholar 

  266. Ashraf S, Nabi M, Rasool SU, Rashid F, Amin S. Hyperandrogenism in polycystic ovarian syndrome and role of CYP gene variants: a review. Egypt J Med Hum. 2019;20:1. doi:https://doi.org/ARTN251186/s43042-019-0031-4.

  267. Williams T, Mortada R, Porter S. Diagnosis and treatment of polycystic ovary syndrome. Am Fam Physician. 2016;94(2):106–13. https://www.ncbi.nlm.nih.gov/pubmed/27419327.

    PubMed  Google Scholar 

  268. Rudnicka E, Suchta K, Grymowicz M, Calik-Ksepka A, Smolarczyk K, Duszewska AM, et al. Chronic low Grade inflammation in Pathogenesis of PCOS. Int J Mol Sci. 2021;22:7. https://doi.org/10.3390/ijms22073789.

    Article  CAS  Google Scholar 

  269. Malkin CJ, Pugh PJ, Jones RD, Kapoor D, Channer KS, Jones TH. The effect of testosterone replacement on endogenous inflammatory cytokines and lipid profiles in hypogonadal men. J Clin Endocrinol Metab. 2004;89:7:3313–8. https://doi.org/10.1210/jc.2003-031069.

    Article  CAS  PubMed  Google Scholar 

  270. Lee H-R, Kim T-H, Choi K-C. Functions and physiological roles of two types of estrogen receptors, ERalpha and ERbeta, identified by estrogen receptor knockout mouse. Lab Anim Res. 2012;28:2. https://doi.org/10.5625/lar.2012.28.2.71.

    Article  Google Scholar 

  271. Villa A, Rizzi N, Vegeto E, Ciana P, Maggi A. Estrogen accelerates the resolution of inflammation in macrophagic cells. Sci Rep. 2015;5:14. https://doi.org/10.1038/srep15224.

    Article  CAS  Google Scholar 

  272. Nakamura TJ, Sellix MT, Menaker M, Block GD. Estrogen directly modulates circadian rhythms of PER2 expression in the uterus. Am J Physiol-Endocrinol Metab. 2008;295(5):E1025–31. https://doi.org/10.1152/ajpendo.90392.2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Chen Q, Zhang W, Sadana N, Chen X. Estrogen receptors in pain modulation: cellular signaling. Biology Sex Differences. 2021;12:1. https://doi.org/10.1186/s13293-021-00364-5.

    Article  CAS  Google Scholar 

  274. Evrard HC. Estrogen synthesis in the spinal dorsal horn: a new central mechanism for the hormonal regulation of pain. Am J Physiol-Regul Integr Comp Physiol. 2006;291(2):R291–9. https://doi.org/10.1152/ajpregu.00930.2005.

    Article  CAS  PubMed  Google Scholar 

  275. Li L, Fan XT, Warner M, Xu XJ, Gustafsson JA, Wiesenfeld-Hallin Z. Ablation of estrogen receptor alpha or beta eliminates sex differences in mechanical pain threshold in normal and inflamed mice. Pain. 2009;143(1–2):37–40. https://doi.org/10.1016/j.pain.2009.01.005.

    Article  CAS  PubMed  Google Scholar 

  276. Coulombe MA, Spooner MF, Gaumond I, Carrier JC, Marchand S. Estrogen receptors beta and alpha have specific pro- and anti-nociceptive actions. Neuroscience. 2011;184:172–82. https://doi.org/10.1016/j.neuroscience.2011.02.057.

    Article  CAS  PubMed  Google Scholar 

  277. Zhong YQ, Li KC, Zhang X. Potentiation of excitatory transmission in substantia gelatinosa neurons of rat spinal cord by inhibition of estrogen receptor alpha. Mol Pain. 2010;6:5. https://doi.org/10.1186/1744-8069-6-92.

    Article  CAS  Google Scholar 

  278. Tang B, Ji YP, Traub RJ. Estrogen alters spinal NMDA receptor activity via a PKA signaling pathway in a visceral pain model in the rat. Pain. 2008;137:3:540–9. https://doi.org/10.1016/j.pain.2007.10.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, et al. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 2007;87:3905–31. https://doi.org/10.1152/physrev.00026.2006.

    Article  CAS  Google Scholar 

  280. Shughrue PJ, Scrimo PJ, Merchenthaler I. Evidence for the colocalization of estrogen receptor-beta mRNA and estrogen receptor-alpha immunoreactivity in neurons of the rat forebrain. Endocrinology. 1998;139:12:5267–70. https://doi.org/10.1210/en.139.12.5267.

    Article  CAS  PubMed  Google Scholar 

  281. Vrtačnik P, Ostanek B, Mencej-Bedrač S, Marc J. The many faces of estrogen signaling. Biochemia Med. 2014;24:3329–42. https://doi.org/10.11613/bm.2014.035.

    Article  Google Scholar 

  282. Smith YR, Stohler CS, Nichols TE, Bueller JA, Koeppe RA, Zubieta JK. Pronociceptive and antinociceptive effects of estradiol through endogenous opioid neurotransmission in women. J Neurosci. 2006;26:21:5777–85. https://doi.org/10.1523/jneurosci.5223-05.2006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  283. Herrera AY, Mather M. Actions and interactions of estradiol and glucocorticoids in cognition and the brain: implications for aging women. Neurosci Biobehav Rev. 2015;55:36–52. https://doi.org/10.1016/j.neubiorev.2015.04.005.

    Article  CAS  PubMed Central  Google Scholar 

  284. Wu Y-W, Bi Y-P, Kou X-X, Xu W, Ma L-Q, Wang K-W, et al. 17- -Estradiol enhanced Allodynia of Inflammatory Temporomandibular Joint through Upregulation of hippocampal TRPV1 in Ovariectomized rats. J Neurosci. 2010;30:26:8710–9. https://doi.org/10.1523/jneurosci.6323-09.2010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Yamagata K, Sugimura M, Yoshida M, Sekine S, Kawano A, Oyamaguchi A, et al. Estrogens Exacerbate Nociceptive Pain via Up-Regulation of TRPV1 and ANO1 in trigeminal primary neurons of female rats. Endocrinology. 2016;157:11:4309–17. https://doi.org/10.1210/en.2016-1218.

    Article  CAS  PubMed  Google Scholar 

  286. Paredes S, Cantillo S, Candido KD, Knezevic NN. An Association of Serotonin with Pain disorders and its modulation by Estrogens. Int J Mol Sci. 2019;20:225729. https://doi.org/10.3390/ijms20225729.

    Article  CAS  Google Scholar 

  287. Mulak A, Tache Y, Larauche M. Sex hormones in the modulation of irritable bowel syndrome. World J Gastroenterol. 2014;20:10:2433–48. https://doi.org/10.3748/wjg.v20.i10.2433.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Shah NM, Lai PF, Imami N, Johnson MR. Progesterone-related Immune modulation of pregnancy and labor. Front Endocrinol. 2019;10:19. https://doi.org/10.3389/fendo.2019.00198.

    Article  Google Scholar 

  289. Oettel M, Mukhopadhyay AK. Progesterone: the forgotten hormone in men? Aging Male. 2004;7:3236–57. https://doi.org/10.1080/13685530400004199.

    Article  CAS  Google Scholar 

  290. Hall OJ, Klein SL. Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Mucosal Immunol. 2017;10:5. https://doi.org/10.1038/mi.2017.35.

    Article  CAS  Google Scholar 

  291. Arruvito L, Giulianelli S, Flores AC, Paladino N, Barboza M, Lanari C, et al. NK cells expressing a progesterone receptor are susceptible to progesterone-induced apoptosis. J Immunol. 2008;180:8. https://doi.org/10.4049/jimmunol.180.8.5746.

    Article  Google Scholar 

  292. Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16:10. https://doi.org/10.1038/nri.2016.90.

    Article  CAS  Google Scholar 

  293. Butts CL, Shukair SA, Duncan KM, Bowers E, Horn C, Belyavskaya E, et al. Progesterone inhibits mature rat dendritic cells in a receptor-mediated fashion. Int Immunol. 2007;19:3287–96. https://doi.org/10.1093/intimm/dxl145.

    Article  CAS  Google Scholar 

  294. Jones LA, Kreem S, Shweash M, Paul A, Alexander J, Roberts CW. Differential modulation of TLR3- and TLR4-mediated dendritic cell maturation and function by progesterone. J Immunol. 2010;185:8:4525–34. https://doi.org/10.4049/jimmunol.0901155.

    Article  CAS  PubMed  Google Scholar 

  295. Lei BL, Mace B, Dawson HN, Warner DS, Laskowitz DT, James ML. Anti-inflammatory effects of Progesterone in Lipopolysaccharide-stimulated BV-2 microglia. PLoS ONE. 2014;9:7. ; doi:https://doi.org/ARTNe1039691371/journal.pone.0103969.

    Article  Google Scholar 

  296. Shufelt CL, Merz CNB. Contraceptive hormone Use and Cardiovascular Disease. J Am Coll Cardiol. 2009;53:3221–31. https://doi.org/10.1016/j.jacc.2008.09.042.

    Article  CAS  Google Scholar 

  297. Allais G, Chiarle G, Sinigaglia S, Airola G, Schiapparelli P, Bergandi F, et al. Treating migraine with contraceptives. Neurol Sci. 2017;38:S85–9. https://doi.org/10.1007/s10072-017-2906-9.

    Article  Google Scholar 

  298. Matera L. Endocrine, paracrine and autocrine actions of prolactin on immune cells. Life Sci. 1996;59:8:599–614. https://doi.org/10.1016/0024-3205(96)00225-1.

    Article  CAS  PubMed  Google Scholar 

  299. De Bellis A, Bizzarro A, Pivonello R, Lombardi G, Bellastella A. Prolactin and autoimmunity. Pituitary. 2005;8:1:25–30. https://doi.org/10.1007/s11102-005-5082-5.

    Article  CAS  PubMed  Google Scholar 

  300. Gorvin CM. The prolactin receptor: diverse and emerging roles in pathophysiology. J Clin Transl Endocrinol. 2015;2:3:85–91. https://doi.org/10.1016/j.jcte.2015.05.001.

    Article  Google Scholar 

  301. Ben-Jonathan N, Mershon JL, Allen DL, Steinmetz RW. Extrapituitary prolactin: distribution, regulation, functions, and clinical aspects. Endocr Rev. 1996;17:6. https://doi.org/10.1210/edrv-17-6-639.

    Article  Google Scholar 

  302. Mason BN, Kallianpur R, Price TJ, Akopian AN, Dussor GO. Prolactin signaling modulates stress-induced behavioral responses in a preclinical mouse model of migraine. Headache. 2022;62:1:11–25. https://doi.org/10.1111/head.14248.

    Article  PubMed  Google Scholar 

  303. Gazerani P. A link between migraine and prolactin: the way forward. Future Sci OA. 2021;7. https://doi.org/10.2144/fsoa-2021-0047. 9:Fso748.

  304. Bussone G, Usai S, Moschiano F. How to investigate and treat: Headache and Hyperprolactinemia. Curr Pain Headache Rep. 2012;16:4365–70. https://doi.org/10.1007/s11916-012-0267-x.

    Article  Google Scholar 

  305. Strebel PM, Zacur HA, Gold EB, Headache, hyperprolactinemia andprolactinomas. Obstet Gynecol. 1986;68:2:195–9. ://WOS:A1986D432000011.

    CAS  PubMed  Google Scholar 

  306. Bosco D, Belfiore A, Fava A, De Rose M, Plastino M, Ceccotti C, et al. Relationship between high prolactine levels and migraine attacks in patients with microprolactinoma. J Headache Pain. 2008;9:2. https://doi.org/10.1007/s10194-008-0016-z.

    Article  CAS  Google Scholar 

  307. Noori-Zadeh A, Karamkhani M, Seidkhani-Nahal A, Khosravi A, Darabi S. Evidence for hyperprolactinemia in migraineurs: a systematic review and meta-analysis. Neurol Sci. 2020;41:1. https://doi.org/10.1007/s10072-019-04035-7.

    Article  Google Scholar 

  308. Avona A, Mason BN, Burgos-Vega C, Hovhannisyan AH, Belugin SN, Mecklenburg J, et al. Meningeal CGRP-Prolactin Interaction evokes female-specific migraine behavior. Ann Neurol. 2021;89:6:1129–44. https://doi.org/10.1002/ana.26070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Shelly S, Boaz M, Orbach H. Prolactin and autoimmunity. Autoimmun Rev. 2012;11:6–7. https://doi.org/10.1016/j.autrev.2011.11.009.

    Article  CAS  Google Scholar 

  310. Patil M, Belugin S, Mecklenburg J, Wangzhou A, Paige C, Barba-Escobedo PA, et al. Prolactin regulates Pain responses via a female-selective nociceptor-specific mechanism. iScience. 2019;20:449–65. https://doi.org/10.1016/j.isci.2019.09.039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Mecklenburg J, Wangzhou A, Hovhannisyan AH, Barba-Escobedo P, Shein SA, Zou Y, et al. Sex-dependent pain trajectories induced by prolactin require an inflammatory response for pain resolution. Brain Behav Immun. 2022;101:246–63. https://doi.org/10.1016/j.bbi.2022.01.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Paige C, Barba-Escobedo PA, Mecklenburg J, Patil M, Goffin V, Grattan DR, et al. Neuroendocrine mechanisms governing sex differences in Hyperalgesic Priming Involve Prolactin Receptor Sensory Neuron Signaling. J Neurosci. 2020;40:37:7080–90. https://doi.org/10.1523/JNEUROSCI.1499-20.2020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Lin Y, Wang Y, Li PF, PPARα. An emerging target of metabolic syndrome, neurodegenerative and cardiovascular diseases. Front Endocrinol (Lausanne). 2022;13:1074911. https://doi.org/10.3389/fendo.2022.1074911.

    Article  PubMed  Google Scholar 

  314. Okine BN, Gaspar JC, Finn DP. PPARs and pain. Br J Pharmacol. 2019;176:10:1421–42. https://doi.org/10.1111/bph.14339.

    Article  CAS  PubMed  Google Scholar 

  315. Toobian D, Ghosh P, Katkar GD. Parsing the role of PPARs in macrophage processes. Front Immunol. 2021;12:783780.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Zhang MA, Rego D, Moshkova M, Kebir H, Chruscinski A, Nguyen H, et al. Peroxisome proliferator-activated receptor (PPAR)alpha and -gamma regulate IFN gamma and IL-17A production by human T cells in a sex-specific way. Proc Natl Acad Sci U S A. 2012;109:24:9505–10. https://doi.org/10.1073/pnas.1118458109.

    Article  PubMed  PubMed Central  Google Scholar 

  317. Barrett AC. Low efficacy opioids: implications for sex differences in opioid antinociception. Exp Clin Psychopharmacol. 2006;14:1:1–11. https://doi.org/10.1037/1064-1297.14.1.1.

    Article  CAS  Google Scholar 

  318. Niesters M, Dahan A, Kest B, Zacny J, Stijnen T, Aarts L, et al. Do sex differences exist in opioid analgesia? A systematic review and meta-analysis of human experimental and clinical studies. Pain. 2010;151:161–8. https://doi.org/10.1016/j.pain.2010.06.012.

    Article  Google Scholar 

  319. Goetz TG, Becker JB, Mazure CM. Women, opioid use and addiction. Faseb J. 2021;35:2. https://doi.org/10.1096/fj.202002125R.

    Article  CAS  Google Scholar 

  320. Mazure CM, Fiellin DA. Women and opioids: something different is happening here. Lancet. 2018;392:10141.

    Article  Google Scholar 

  321. Sorge RE, Totsch SK. Sex differences in Pain. J Neurosci Res. 2017;95:6:1271–81. https://doi.org/10.1002/jnr.23841.

    Article  CAS  PubMed  Google Scholar 

  322. Mogil JS. OPINION sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci. 2012;13:12859–66. https://doi.org/10.1038/nrn3360.

    Article  CAS  Google Scholar 

  323. Serrao M, Rossi P, Sandrini G, Amabile GA, Nappi G, Pierelli F. Effects of diffuse noxious inhibitory controls on temporal summation of the RIII reflex in humans. Pain. 2004;112:3. https://doi.org/10.1016/j.pain.2004.09.018.

    Article  Google Scholar 

  324. Weissman-Fogel I, Sprecher E, Pud D. Effects of catastrophizing on pain perception and pain modulation. Exp Brain Res. 2008;186:1:79–85. https://doi.org/10.1007/s00221-007-1206-7.

    Article  PubMed  Google Scholar 

  325. Vina ER, Ran D, Ashbeck EL, Kwoh CK. Widespread Pain is Associated with increased risk of no clinical improvement after TKA in women. Clin Orthop Rel Res. 2020;478:7:1453–65. https://doi.org/10.1097/corr.0000000000001001.

    Article  Google Scholar 

  326. Patel B, Eskander MA, Fang-Mei Chang P, Chapa B, Ruparel SB, Lai Z, et al. Understanding painful versus non-painful dental pain in female and male patients: a transcriptomic analysis of human biopsies. PLoS ONE. 2023;18:9e0291724. https://doi.org/10.1371/journal.pone.0291724.

    Article  CAS  Google Scholar 

  327. See LP, Sripinun P, Lu W, Li J, Alboloushi N, Alvarez-Periel E, et al. Increased Purinergic Signaling in Human Dental Pulps with Inflammatory Pain is sex-dependent. J Pain. 2024;25:4:1039–58. https://doi.org/10.1016/j.jpain.2023.10.026.

    Article  CAS  PubMed  Google Scholar 

  328. Kriegova E, Manukyan G, Mikulkova Z, Gabcova G, Kudelka M, Gajdos P, et al. Gender-related differences observed among immune cells in synovial fluid in knee osteoarthritis. Osteoarthritis Cartilage. 2018;26:9:1247–56. https://doi.org/10.1016/j.joca.2018.04.016.

    Article  CAS  PubMed  Google Scholar 

  329. Keselman A, Fang X, White PB, Heller NM. Estrogen Signaling Contributes to sex differences in macrophage polarization during Asthma. J Immunol. 2017;199:5:1573–83. https://doi.org/10.4049/jimmunol.1601975.

    Article  CAS  PubMed  Google Scholar 

  330. Li K, Xu W, Guo Q, Jiang Z, Wang P, Yue Y, et al. Differential macrophage polarization in male and female BALB/c mice infected with coxsackievirus B3 defines susceptibility to viral myocarditis. Circ Res. 2009;105:4353–64. https://doi.org/10.1161/CIRCRESAHA.109.195230.

    Article  CAS  Google Scholar 

  331. Becerra-Diaz M, Lerner AD, Yu DH, Thiboutot JP, Liu MC, Yarmus LB, et al. Sex differences in M2 polarization, chemokine and IL-4 receptors in monocytes and macrophages from asthmatics. Cell Immunol. 2021;360:10. https://doi.org/10.1016/j.cellimm.2020.104252.

    Article  CAS  Google Scholar 

  332. Rozen TD, Fishman RS. Female cluster headache in the United States of America: what are the gender differences? Results from the United States Cluster Headache Survey. J Neurol Sci. 2012;317(1–2):17–28. https://doi.org/10.1016/j.jns.2012.03.006.

    Article  PubMed  Google Scholar 

  333. Gutiérrez-Albaladejo N, López-de-Andrés A, Cuadrado-Corrales N, Albaladejo-Vicente R, Villanueva-Orbaiz R, Carricondo F, et al. Asthma is Associated with Back Pain and Migraine-results of Population-based case-control study. J Clin Med. 2023;12:22. https://doi.org/10.3390/jcm12227107.

    Article  Google Scholar 

  334. 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. J Neurosci. 2019;39:22:4323–31. https://doi.org/10.1523/JNEUROSCI.0364-19.2019.

    Article  PubMed  PubMed Central  Google Scholar 

  335. Wattiez AS, Sowers LP, Russo AF. Calcitonin gene-related peptide (CGRP): role in migraine pathophysiology and therapeutic targeting. Expert Opin Ther Targets. 2020;24:2:91–100. https://doi.org/10.1080/14728222.2020.1724285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  336. Deen M, Correnti E, Kamm K, Kelderman T, Papetti L, Rubio-Beltran E, et al. Blocking CGRP in migraine patients - a review of pros and cons. J Headache Pain. 2017;18:196. https://doi.org/10.1186/s10194-017-0807-1.

    Article  CAS  Google Scholar 

  337. Taylor RW, Grant AM, Williams SM, Goulding A. Sex differences in Regional Body Fat distribution from pre- to Postpuberty. Obesity. 2010;18:7:1410–6. https://doi.org/10.1038/oby.2009.399.

    Article  PubMed  Google Scholar 

  338. Berwal D, Branisteanu DD, Glickman M, Sagar A, Pilitsis JG. The sex-dependent impact of adipose tissue and inflammation on chronic pain - A cross-sectional study from the all of us research program. Cytokine. 2024;179:156614. https://doi.org/10.1016/j.cyto.2024.156614.

    Article  CAS  PubMed  Google Scholar 

  339. Cartier A, Cote M, Lemieux I, Perusse L, Tremblay A, Bouchard C, et al. Sex differences in inflammatory markers: what is the contribution of visceral adiposity? Am J Clin Nutr. 2009;89:5:1307–14. https://doi.org/10.3945/ajcn.2008.27030.

    Article  CAS  PubMed  Google Scholar 

  340. Varghese M, Griffin C, McKernan K, Eter L, Lanzetta N, Agarwal D, et al. Sex differences in inflammatory responses to adipose tissue lipolysis in Diet-Induced obesity. Endocrinology. 2019;160:2:293–312. https://doi.org/10.1210/en.2018-00797.

    Article  CAS  PubMed  Google Scholar 

  341. Kim JJ, Sears DD. TLR4 and insulin resistance. Gastroenterol Res Pract. 2010;2010:1–11. https://doi.org/10.1155/2010/212563.

    Article  Google Scholar 

  342. Tramullas M, Finger BC, Dinan TG, Cryan JF. Obesity takes its toll on Visceral Pain: high-Fat Diet induces toll-like receptor 4-Dependent visceral hypersensitivity. PLoS ONE. 2016;11:5e0155367. https://doi.org/10.1371/journal.pone.0155367.

    Article  CAS  Google Scholar 

  343. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116:11:3015–25. https://doi.org/10.1172/jci28898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  344. Vitseva OI, Tanriverdi K, Tchkonia TT, Kirkland JL, McDonnell ME, Apovian CM, et al. Inducible toll-like receptor and NF-kappaB regulatory pathway expression in human adipose tissue. Obes (Silver Spring). 2008;16:5. https://doi.org/10.1038/oby.2008.25.

    Article  CAS  Google Scholar 

  345. Tighe PJ, Riley JL 3rd, Fillingim RB. Sex differences in the incidence of severe pain events following surgery: a review of 333,000 pain scores. Pain Med. 2014;15:81390–404. https://doi.org/10.1111/pme.12498.

  346. Bjørnnes AK, Parry M, Lie I, Fagerland MW, Watt-Watson J, Rustøen T, et al. Pain experiences of men and women after cardiac surgery. J Clin Nurs. 2016;25:19–20. https://doi.org/10.1111/jocn.13329.

    Article  Google Scholar 

  347. Chung J, Stevens LM, Ouzounian M, El-Hamamsy I, Bouhout I, Dagenais F, et al. Sex-related differences in patients undergoing thoracic aortic surgery. Circulation. 2019;139:9:1177–84. https://doi.org/10.1161/circulationaha.118.035805.

    Article  PubMed  Google Scholar 

  348. Moscarelli M, Lorusso R, Angelini GD, Di Bari N, Paparella D, Fattouch K, et al. Sex-specific differences and postoperative outcomes of minimally invasive and sternotomy valve surgery. Eur J Cardiothorac Surg. 2022;61:3:695–702. https://doi.org/10.1093/ejcts/ezab369.

    Article  Google Scholar 

  349. Nandi M, Schreiber KL, Martel MO, Cornelius M, Campbell CM, Haythornthwaite JA, et al. Sex differences in negative affect and postoperative pain in patients undergoing total knee arthroplasty. Biol Sex Differ. 2019;10:123. https://doi.org/10.1186/s13293-019-0237-7.

    Article  Google Scholar 

  350. Dos Santos NL, Lenert ME, Castillo ZW, Mody PH, Thompson LT, Burton MD. Age and sex drive differential behavioral and neuroimmune phenotypes during postoperative pain. Neurobiol Aging. 2023;123:129–44. https://doi.org/10.1016/j.neurobiolaging.2022.09.008.

    Article  PubMed  Google Scholar 

  351. Verriotis M, Jones L, Whitehead K, Laudiano-Dray M, Panayotidis I, Patel H, et al. The distribution of pain activity across the human neonatal brain is sex dependent. NeuroImage. 2018;178:69–77. https://doi.org/10.1016/j.neuroimage.2018.05.030.

    Article  PubMed  Google Scholar 

  352. Tan JS, Tikoft E, O’Sullivan P, Smith A, Campbell A, Caneiro JP, Kent P. The relationship between changes in movement and activity limitation or pain in people with knee osteoarthritis: a systematic review. J Orthop Sports Phys Ther. 2021;1:10. https://doi.org/10.2519/jospt.2021.10418

  353. Astephen Wilson JL, Dunbar MJ, Hubley-Kozey CL. Knee joint biomechanics and neuromuscular control during gait before and after total knee arthroplasty are sex-specific. J Arthroplasty. 2015;30:1. https://doi.org/10.1016/j.arth.2014.07.028.

    Article  Google Scholar 

  354. Valdrighi N, Vago JP, Blom AB, van de Loo FAJ, Blaney Davidson EN. Innate immunity at the core of sex differences in Osteoarthritic Pain? Front Pharmacol. 2022;13:881500. https://doi.org/10.3389/fphar.2022.881500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  355. Mun CJ, Letzen JE, Nance S, Smith MT, Khanuja HS, Sterling RS, et al. Sex differences in Interleukin-6 responses over Time following Laboratory Pain Testing among patients with knee osteoarthritis. J Pain. 2020;21:5–6. https://doi.org/10.1016/j.jpain.2019.11.003.

    Article  CAS  Google Scholar 

  356. Kim JR, Kim HA. Molecular mechanisms of sex-related differences in arthritis and Associated Pain. Int J Mol Sci. 2020;21:21. https://doi.org/10.3390/ijms21217938.

    Article  CAS  Google Scholar 

  357. Pan Q, O’Connor MI, Coutts RD, Hyzy SL, Olivares-Navarrete R, Schwartz Z, et al. Characterization of osteoarthritic human knees indicates potential sex differences. Biol Sex Differ. 2016;7:27. https://doi.org/10.1186/s13293-016-0080-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  358. Kosek E, Finn A, Ultenius C, Hugo A, Svensson C, Ahmed AS. Differences in neuroimmune signalling between male and female patients suffering from knee osteoarthritis. J Neuroimmunol. 2018;321:49–60. https://doi.org/10.1016/j.jneuroim.2018.05.009.

    Article  CAS  PubMed  Google Scholar 

  359. Solheim N, Östlund S, Gordh T, Rosseland LA. Women report higher pain intensity at a lower level of inflammation after knee surgery compared with men. Pain Rep. 2017;2:3e595. https://doi.org/10.1097/pr9.0000000000000595.

    Article  Google Scholar 

  360. Perruccio AV, Badley EM, Power JD, Canizares M, Kapoor M, Rockel J, et al. Sex differences in the relationship between individual systemic markers of inflammation and pain in knee osteoarthritis. Osteoarthr Cartil Open. 2019;1(1–2):100004. https://doi.org/10.1016/j.ocarto.2019.100004.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank current and past Burton lab members for aiding in the development of this manuscript. All graphics were generated by BioRender.com. The content of this manuscript (portions of text) has previously appeared in the thesis of Thomas A. Szabo-Pardi, available online.

Funding

This research was funded by the NIH/NIDDK, grant number R21DK130015 (M.D.B.), NIH/NIGMS, grant number R35GM147094, the University of Texas System STARS program research support grant (M.D.B.), and the Rita Allen Foundation Grant (M.D.B.).

Author information

Authors and Affiliations

Authors

Contributions

All authors were involved with the conception of the manuscript. S.N.A., A.R.G., E.K.D., & L.R.F. contributed to all phases of writing, research, and bibliography development. A.R.G. developed the initial draft of the figures, H.M.K.A. and T.S.P. contributed to initial drafts and final editing of the manuscript. M.D.B. participated and supervised in all aspects from conception to manuscript preparation, and funding acquisition.

Corresponding author

Correspondence to Michael D. Burton.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Competing interests

The authors (Alexander, S.N, Green, A.R., Debner, E.K., Abdelhadi, H.M.K., Ramos Freitas, L.E., Szabo-Pardi, T.A., Burton, M.D) have no conflict of interests to declare.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Shevon N. Alexander, Audrey R. Green, Emily K. Debner and Lindsey E. Ramos Freitas are co-first authors.

Glossary

At-HMGB1

All-thiol form HMGB1

CGRP

Calcitonin gene-related peptide

CIPN

Chemotherapy-induced peripheral neuropathy

CSF1

Colony-stimulating factor 1

DAMPs

Damage-associated molecular patterns

DNIC

Diffuse noxious inhibitory controls

DRG

Dorsal root ganglion

Ds-HMGB1

Disulfide HMGB1

E2

Estradiol

ER

Estrogen receptor

Fc

Receptors for immunoglobulins

HMGB1

High mobility group box 1

LPS

Lipopolysaccharide

M1/2 polarization

Pro-inflammatory (M1) and anti-inflammatory (M2) states

Nav

Voltage-gated sodium channel

NK

Natural killer cell

PAMPs

Pathogen-associated molecular patterns

PCOS

Polycystic ovary syndrome

PPARs

Peroxisome proliferator-activated receptor

PRLR

Prolactin receptor

SABV

Sex as a biological variable

TG

Trigeminal ganglion

TLR

Toll-like receptor

TNF-α

Tumor necrosis factor alpha

TRPA1

Transient receptor potential ankyrin 1

TRPV1

Transient receptor potential vanilloid 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alexander, S.N., Green, A.R., Debner, E.K. et al. The influence of sex on neuroimmune communication, pain, and physiology. Biol Sex Differ 15, 82 (2024). https://doi.org/10.1186/s13293-024-00660-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13293-024-00660-w

Keywords