Skip to main content

REVIEW article

Front. Immunol., 13 March 2017
Sec. Multiple Sclerosis and Neuroimmunology
This article is part of the Research Topic Neuroimmune interface in health and diseases View all 15 articles

Lifetime Modulation of the Pain System via Neuroimmune and Neuroendocrine Interactions

  • 1Laboratory for Molecular Mechanisms of Thalamus Development, RIKEN BSI, Wako, Japan
  • 2Department of Clinical Neuroscience, Division for Psychology, Karolinska Institutet, Solna, Sweden
  • 3Stress Research Institute, Stockholm University, Stockholm, Sweden

Chronic pain is a debilitating condition that still is challenging both clinicians and researchers. Despite intense research, it is still not clear why some individuals develop chronic pain while others do not or how to heal this disease. In this review, we argue for a multisystem approach to understand chronic pain. Pain is not only to be viewed simply as a result of aberrant neuronal activity but also as a result of adverse early-life experiences that impact an individual’s endocrine, immune, and nervous systems and changes which in turn program the pain system. First, we give an overview of the ontogeny of the central nervous system, endocrine, and immune systems and their windows of vulnerability. Thereafter, we summarize human and animal findings from our laboratories and others that point to an important role of the endocrine and immune systems in modulating pain sensitivity. Taking “early-life history” into account, together with the past and current immunological and endocrine status of chronic pain patients, is a necessary step to understand chronic pain pathophysiology and assist clinicians in tailoring the best therapeutic approach.

Introduction

The pain system is modulated by neuroimmune and neuroendocrine mechanisms from embryonic development throughout life. Unlike the traditional reductionist view that posits that pain is solely due to aberrant spinal and supraspinal neuronal activity, we now understand pain in the context of a complex multisystem comprising well-organized interactions between neuroendocrine and neuroimmune systems (1). The changes in the nervous system induced by the immune system and the endocrine system are of both structural and functional character and are a part of the normal, adaptive development of the pain system. However, an adaptation that is advantageous in one situation may pose a risk factor in another. Exposure to a wide range of stressors, from physical injury (such as incision) to infection and inflammation [as induced by, e.g., lipopolysaccharide (LPS)], activates the hypothalamo–pituitary–adrenal (HPA) axis as well as peripheral and central immune responses and reorganizes the sensitivity of the pain system (25). The HPA axis and neuroimmune activation are of importance in determining long-term pathological states such as chronic pain.

Treating chronic pain is complicated by the wide individual differences in symptoms and treatment response. Chronic pain is also associated with a high incidence of psychiatric comorbidity (6) and is often present with other primary diagnoses, such as inflammatory disease. Furthermore, stress is often directly targeted in behavioral treatment strategies for chronic pain (7), as part of an integrated treatment approach (8, 9). In this study, we explore some of the biological mechanisms that may form the foundations of the complexity seen in clinical pain.

This review focuses on some of the mechanisms involved in the maturation of the nervous system, which define the function of the pain system later in life. We highlight the importance of neuroimmune and neuroendocrine interactions very early in life in the programming of the pain system. We also discuss how the immune system and the endocrine system continue to modulate pain processing throughout life and about the significance of these interactions for chronic pain.

Ontogeny of the Central Nervous System (CNS) During the Prenatal and Postnatal Period

Neuronal circuits are forged by sensory experiences. Exposure to environmental stressors during a critical period of brain ontogeny, when neuronal circuits are particularly sensitive to modification by experience, can have long-term consequences on neural circuits, ultimately affecting behavior (10). Although our genetic makeup determines much of the structure and function of the nervous system, the environment where the individual is born, as well as the environmental conditions that will accompany the individual throughout his/her life, plays a crucial role in tailoring the neuronal properties. The postnatal developing nervous system responds to the external world to shape its neural circuits in order to subserve a particular function (i.e., vision, auditory, touch, etc.). In normal conditions (i.e., in the absence of any adverse events), non-stressful early experience specifies a neural trajectory to the best possible circuits of connectivity. In other words, non-efficient connections are eliminated and those that are functionally stable remain. However, if exposed to stress—whether it is of physical, physiological, psychological, or viral/bacterial nature—during a time when the brain is still undergoing fine-tuned maturation, the process of synaptic plasticity, or synaptic tuning can go seriously wrong, affecting the behavioral outcome.

Early Development of the Human Brain

During the prenatal period, the brain produces approximately 250,000 cells per minute (11). Neuronal migration occurs between gestational week (GW) 8 and 16 forming the subventricular zone (SVZ) (12). Around GW 16, neurons reach their final target and begin to form connections among brain regions (13). Synapse formation in both the auditory and prefrontal cortices begins around GW 27 (14). During the beginning of the third trimester, synaptogenesis occurs with a rate of approximately 40,000 synapses per minute (15). Subsequently, myelination as well as proliferation and differentiation of oligodendrocytes (cells that produce myelin) take place. After birth, the size of the brain continues to increase dramatically, with intense metabolic changes associated with synapse formation and axonal growth during the first 3 months of postnatal life (16). The way the complex human brain develops and matures is through a significant increase in volume due to overproduction of synapses, myelination, and connections during infancy, followed by the elimination of less efficient synapses via pruning (17). Most importantly, the developmental trajectory of the neocortex is different depending on brain regions. For instance, the primary visual cortex undergoes significant maturation during the first 3 months of life, whereas the primary auditory cortex continues to mature over the first 3 years of life (18). The bilateral thalamic connectivity to the prefrontal cortex (PFC) is increased gradually from childhood to late teens (19), and synaptic pruning in the PFC continues to occur in mid-adolescence (14). The relatively late maturation of thalamo–PFC synaptic connections implies that key connections involved in complex cognitive functions, including pain, are still undergoing fine-tuned maturation in early postnatal life. Consequently, exposure to stressful events such as viral/bacterial infections during postnatal life is likely to be able to alter key neural circuits involved in pain processing. This may lead to altered pain responses later in life. At present, there is a paucity of research tackling this question, and further studies investigating the impact of early-life stress on neural circuits involved in pain processing are needed.

What Animal Models Reveal about Neurogenesis and Synaptic Plasticity

The traditional dogma posits that the postnatal brain (including adult brain) possesses a fixed number of neurons that are generated from birth and that no neurogenesis or synaptic plasticity is possible in the adult brain (20). However, it is now clear that neurogenesis and synaptic plasticity continue to occur in the adult brain, although at a lower rate. Findings from studies that used standard neuronal markers, such as NeuN and bromodeoxyuridine (BrdU), have detected postnatal neurogenesis both in primates and rodents. NeuN+/BrdU+ cells were detected particularly in two regions: the SVZ–olfactory bulb and the subgranular zone (SGZ)–hippocampal granule cell layer (2125). Regarding synaptic plasticity in the adult brain, pioneer studies by Merzenich et al. demonstrated that amputation of one finger in adult monkeys resulted in deafference of the devoted territory within the somatosensory cortex and that this region compensated by receiving inputs from neighboring fingers (26). Later on, Robertson and Irvine showed that similar compensatory mechanisms and cortical rearrangement occurred in the auditory cortex following lesion of the cochlea (27).

In rats, PFC neural circuits undergo significant changes during the perinatal period. The myelination of the medial PFC (mPFC) is very low at P7, increases gradually over the period P21–P50, and reaches peak level at P90 (28). The ontogenic development of the PFC implies that this region, which plays a critical role in cognitive functioning and pain processing (29), is particularly susceptible to environmental stimuli during the neonatal period. Consequently, exposure to stressful events during this period is likely to alter the neural circuits within the PFC—and consequently pain processing later in life. Indeed, sensory, painful, or stressful experience has been shown to change the dendritic and spine morphology in this area. A combination of prenatal stress (E14–E21) and maternal separation (P2–P21) resulted in increased c-Fos expression in the mPFC and reduced dendritic length and dendritic spines of mPFC neurons (30). A recent study has found that pyramidal neurons from the mPFC of spared nerve injury (SNI) rats are characterized by longer basal dendrites and increased spine density compared to sham-operated animals (29). Electrophysiological recording of mPFC pyramidal neurons from SNI rats revealed increased NMDA/AMPA ratio in currents evoked by stimulation of layer 5 (29). However, convincing data linking directly altered PFC neural circuits following early-life stress to future pain responses are still lacking.

Taken together, these data suggest that the perinatal, up to and including the early childhood period, is a time of high plasticity for the brain and adverse events occurring during this critical period of cellular proliferation, differentiation, and maturation can interfere with the normal developmental trajectory of the brain, resulting in structural and/or functional changes in cells, tissues, or organ systems. These changes are proposed to potentially lead to increased susceptibility to neurodevelopmental disorders in later life (3133) and may also be critical for determining adult pain responses and potentially the susceptibility to develop chronic pain.

Early-Life Development of the Pain System

One of the neuronal systems that undergo significant malleability during the perinatal period is the nociceptive system. For instance, at embryonic day (E) 15–17 myelinated A fibers are the first to penetrate the spinal lumbar cord before the subsequent projection of C fibers into the substantia gelatinosa (lamina II, superficial dorsal horn that contains nociceptive-specific neurons) at E19 (34). During the neonatal period, lamina II is innervated by both A- and C fibers. During the first 3 weeks of postnatal age, a withdrawal of A fiber primary afferents into deeper laminae is noticed, and C fibers exclusively innervate lamina II at the adult stage (35). This developmental pattern of nociceptive fibers is of particular relevance to the concept that early-life insults are able to alter the neuroanatomical components of nociception (including nociceptive fibers), leading to altered pain responses later in life. For instance, skin wound during the neonatal period is associated with hyperinnervation of the wounded area by both Aδ and C fibers (36, 37). This hyperinnervation of nociceptive fibers can lead to peripheral sensitization and increased pain sensitivity (i.e., hyperalgesia). Despite the apparent lack of maturity of the nociceptive system, overall, younger animals are markedly more sensitive to noxious stimuli than their adult counterparts (38). Their behavioral output may, however, differ from adult animals. The withdrawal threshold from heat stimuli is lower in young animals compared to adults, and neonatal rats are significantly more (i.e., 10-fold higher) sensitive to formalin injection than preadolescent rats who require higher formalin doses to elicit the formalin-induced behavioral responses (38). For example, until P10, injection of formalin into the hind paw elicits predominantly non-specific whole body movement (i.e., jerking), whereas the formalin-induced specific behaviors such as hind paw shaking, flexion, and licking appears only after P10 (39). Of particular interest, recent studies predominantly from Hathway et al. elegantly demonstrated that the descending inhibitory control of spinal nociceptive reflexes from the periaqueductal gray (PAG) to rostroventral medullar (RVM) in rats undergoes an important developmental switch from facilitatory in young rats to inhibitory in adult rats (4042). This developmental switch was found to be driven by opioid actions on RVM, as microinjection of the μ-opioid agonist [d-Ala2, N-MePh4, glycol]-enkephalin (DAMGO) into RVM facilitates spinal nociceptive reflexes in preadolescent rats (P21), but elicited antinociceptive actions in adult rats (42), and similar response pattern has also been recently shown to occur at the PAG level (41).

Overall, a number of neural systems, including those involved in pain modulation, are characterized by significant malleability illustrated by major structural and functional rearrangements in neural circuits following insult. This injury-induced plasticity renders the nociceptive system more vulnerable to future challenges. Why certain patients develop chronic pain while others do not might in fact result from different early-life experiences in these patients, which may have programmed the pain system differently later in life. Therefore, taking “early-life history” into account is a necessary step to understand chronic pain pathophysiology and developing individual-based therapeutic strategies (43).

Ontogeny of the HPA Axis During the Perinatal Period

Prenatal Development

The experience of stress, from an evolutionary perspective, is very important in promoting survival of an organism. A fundamental system that is subjected to programming by early-life events is the neuroendocrine axis that mediates the stress response, the HPA axis (44, 45). Activation of this system starts with the recruitment of neurons within the paraventricular nucleus of the hypothalamus (PVN), and the end product is the release from the adrenal cortex of corticosterone for rodents or cortisol for humans, via the release of corticotropin-releasing hormone (CRH) and adrenocorticotropic hormone (ACTH) [the HPA axis has been extensively reviewed elsewhere, please see Ref. (46)]. During pregnancy, there is an increase in CRH production in the placenta and fetal membranes. The gradual increase in maternal HPA axis activity during this period leads to maternal hypercortisolemia (47, 48). The fetus has much lower levels of glucocorticoids than its mother although endogenous glucocorticoids can cross the placenta easily. A total of 10–20% of cortisol present in the amniotic liquid is from maternal origin, while the remaining 80–90% gets converted into inactive cortisone by an enzyme, 11β-HSD2, to protect the fetus’s brain from excess glucocorticoids, which can be neurotoxic (49). During the third trimester, fetal 11β-HSD2 levels decrease, and the fetus is exposed to high levels of CRH and cortisol. This rise in CRH and cortisol levels is thought to play an important role in the maturation of organs and preparation of the fetus to the ex utero environment (50). The hippocampus plays a key role in regulating homeostatic levels of glucocorticoids under conditions of stress, and CRF has been shown to modulate the electrical activity of hippocampal neurons (51). Glucocorticoid receptor (GR) mRNAs were detected in human fetal hippocampus at 24 GWs (52). Additionally, fibers expressing CRH have been detected in humans by GW16 (53), and the release of CRH into the pituitary has been reported to occur at GW11.5 (54). At the pituitary, a basic adenohypophysis can be detected at GW6 (55), and by GW8 the pituitary reaches mature stage and can release ACTH (56).

In rodents, GR mRNA can be detected in the telencephalon as early as E12.5 with high expression seen in the anterior hypothalamus, pons, spinal cord, and pituitary gland (57). At E14.5, the expression of GR mRNA significantly increases in the ventral spinal cord and the thalamus and undergoes a moderate decrease in these regions by E15.5. An increase in GR mRNA levels is observed at the same time point in other regions including the neocortex, cerebellum, and basal ganglia (57). In the PVN, GR mRNA can be visible at E16, although it is not clear whether this PVN GR is functional at this stage (58). During the late gestation (E17–E19), GR mRNA is localized in the hippocampus, thalamus, and the amygdala (58). Mineralocorticoid receptors (MR) ontogenetic expression, however, follows a different pattern in rodents. MR mRNA expression cannot be detected before E15.5 when a moderate expression is observed in pituitary gland, brain stem, tegmentum, and neuroepithelium of the septum and pallidum (57). MR mRNA expression is first seen in the hypothalamus at E17.5 and by E19.5 there is a dramatic increase in MR mRNA expression in the hippocampus, septum, anterior hypothalamus, PAG area, and brainstem neuroepithelium (57). Regarding the ontogeny of 11β-HSD2 mRNA (encoding an enzyme that converts corticosterone into its inactive form) in rodents, the expression of 11β-HSD2 mRNA is observed at E11.5 on hippocampal and subicular regions, neocortex, septum, and posterior hypothalamic area. At E14.5, the expression intensity of 11β-HSD2 mRNA starts to decline in the neocortex, pallidal area, and spinal cord, and by E15.5 11β-HSD2 mRNA is restricted to the thalamus, midbrain, striatum, cerebellum, hypothalamus, medulla, and pallidum (57).

Postnatal Modulation

There is a particular period called “the stress hyporesponsive period” (SHRP) from P4 to P14 in rats and from P2 to P12 in mice during which corticosterone levels, as well as ACTH, are maintained at low levels even in the presence of mild stress (59). Although, it is generally accepted that pups do not respond to stress with an elevated HPA axis activity during the SHRP period, it has been reported that 12 day-old pups that were separated from their mothers for 24 h with no access to food or water showed a significant increase in both basal and stress-induced corticosterone and ACTH secretion (59, 60). These results indicate that the HPA axis is particularly sensitive to maternal care even during the SHRP. During this period, high expression of CRH is observed in the PVN, whereas hippocampal GR expression is low at birth and increases gradually during the SHRP (61). In situ hybridization studies in marmoset showed that the ontogenetic profile of MR and GR is different during the postnatal period. Although GR mRNA expression in the dentate gyrus is higher in 4–6 week-old marmoset than in neonates (P1–P2), juveniles (4–5 months), and adult (3–6 years), MR mRNA expression was developmentally consistent in the hippocampus and PVN throughout life (62).

Although we need to proceed with caution when extrapolating from animal studies to humans, the development of the brain in terms of synapse formation and brain growth rate in a P6 rat is relatively equivalent to 38–40 weeks of gestation in humans (63, 64). For obvious ethical and methodological reasons, human data regarding the ontogenetic development of HPA axis are lacking. However, we can conclude from the abovementioned animal data that the prenatal period together with the first 2 weeks of postnatal life constitute a window of significant plasticity for the neuroendocrine system. Homeostasis of the neuroendocrine function, and consequently any physiological system that is under the influence of this system (e.g., pain), is needed for normal neuroendocrine development. Excessive stress that may challenge or perturb the neuroendocrine system when it is still developing could potentially have far-reaching consequences. This way, early-life stress may alter pain, neuroimmune, and neuroendocrine responses for life (4, 6569).

Early Development of the Immune System

Immaturity of the Neonatal Immune System and Susceptibility to Infection

Infant mortality due to infection is high particularly in developing countries with a high prevalence of infection during the neonatal period (70). This high susceptibility of neonates and preterm infants to infection is thought to be due to immaturity of the neonatal immune system. Analysis of umbilical cord from preterm infants revealed fewer naïve CD8+ T cells and regulatory CD31 expression compared to full-term neonates (71). T cells play an important role in the control of intracellular infections. Both human and murine neonates lack mucosally distributed memory CD8+ T cells. Although T cell and cytokine mRNA levels [i.e., interleukin (IL)-1β, IL-6, and IFN-δ] can be detected in the thymus of mice from GD15 (72), neonatal mouse macrophages do not react in an adequate way early in life. For example, T-cells are characterized by lower IFN-δ responses following stimulation (73, 74). Ex vivo stimulation with the bacterial mimetic LPS in mice produced much less pro-inflammatory and anti-inflammatory cytokines response in neonates compared to adult mice (75). The same trend was observed in a human study whereby neonatal monocytes and dendritic cells produced less tumor necrosis factor (TNF)-alpha, IL-12, and IL-6 following LPS stimulation (76). When stimulated with an anti-CD3 antibody, neonatal T cell proliferation significantly decreased compared to adult T cell proliferation. This attenuation of proliferation in neonatal T cells was restored to adult levels following the addition of exogenous IL-2 (77). Furthermore, the total cell number of T cell subtypes (CD4+, CD8+, and Thy1+) is markedly lower in the spleen and lymphoid nodes in P4 mice compared to adult mice (78). Similarly, the function of antigen-presenting cells (APCs) is markedly decreased in human and murine neonates compared to adults (78). Treatment of both immunocompetent and immunodeficient mice with IL-12, a cytokine produced by APCs (79), prior to inoculation with the parasite Cryptosporidium parvum oocysts markedly reduced the severity of infection (80). Additionally, neonatal mice exhibited reduced levels of peripheral IL-12, and mice treated with IL-12 24 h after birth displayed increased levels of IFN-δ and IL-10 mRNA in the spleen (81). Adult humans exhibited much higher levels of granzyme B+ effector differentiated memory CD8+ T cells, which are thought to be the first responders to infections (82), than human neonates (83).

The incidence of sepsis, defined as a systemic inflammatory condition that occurs following exposure to pathogenic microorganisms or their toxins, is more than 25 times higher in infants less than 1 year compared to children from 1 to 14 years of age and constitutes a major risk of mortality and morbidity in the pediatric population (84). The incidence of infections is particularly high during the first postnatal weeks and rapidly decreases thereafter (85). Common causes of infections in neonates include commensal bacteria such as Escherichia coli (85). Both adaptive and innate neonatal immune responses are relatively immature as indicated by a lack of preexisting memory and decreased Th1-type responses (86, 87) as well as impaired production of TNFα following exposure to LPS (88, 89). Neonatal monocyte dendritic cells (moDC) also showed decreased production of interferon-β (IFN-β) in response to in vitro stimulation with LPS compared to mature adult moDC (89, 90). Additionally, whole blood neutrophil concentrations in 1-month children are shown to be lower than those in adults (91).

This immaturity of the immune system during neonatal life may thus predispose the neonatal immune system to infection, both of intra- and extracellular types. Overall, bacterial infection is considered the number one cause of perinatal infection in newborns worldwide (92, 93), which results in increased infant mortality particularly in developing countries (92, 94). In the coming sections, we argue that this sensitivity of the immune system early in life may have long-lasting effects in the adult organism.

Infection As a Perinatal Stressor

Exposure to pathogens early in life is a common event and is considered to play a crucial role in priming the neuroendocrine–neuroimmune interface (95). An infection may not only be life-threatening to an infant but may also reorganize the function of the nervous system, due to the tight interplay between the nervous and immune systems. Human and animal studies have demonstrated that perinatal exposure to an immune challenge can produce changes in the CNS structure and function, leading to an increased risk of developing behavioral and psychopathological alterations later in life (66, 96100). For instance, offspring from mothers exposed to infections such as influenza, LPS, and viral RNA (Poly I:C) during pregnancy have higher risk of developing schizophrenia and autism (101106). A significant number of human and animal studies have also indicated that perinatal infection can alter immune (97, 107110), metabolic (111, 112), reproductive (113, 114), endocrine (95, 115, 116), neurological (117, 118), and cognitive and behavioral responses later in life (98, 119, 120). Interestingly, exposure to LPS in rodents and humans can also cause pain facilitation such as thermal hyperalgesia, mechanical allodynia, and hyperalgesia (121125). Such behavioral findings appear to be the result of altered peripheral and central cytokine activity (122, 126128). Increased levels of pro-inflammatory cytokines, including IL-1β, TNF-α, and IL-6 produced by the maternal or fetal immune system, have been linked to abnormal brain development and increased risk of developing psychopathology (96, 98100). Moreover, higher amounts of IL-6 in the amniotic fluid following bacterial infection during pregnancy have been previously reported to strongly correlate with increased mortality rates and brain injury (129).

Taken together, these findings highlight the fundamental role of the microbial environment in programming behavioral and neural responses. In order to understand the mechanisms of perinatal neuroendocrine–neuroimmune interaction, researchers employ experimental models that mimic the antigenic actions of infection.

LPS as an Experiemental Immunological Stressor

Lipopolysaccharide, a complex glycolipid that is the major component of Gram-negative cell wall usually derived from Salmonella enteritidis or E. coli, is a powerful activator of innate immune responses and induces behavioral symptomatology in the host largely identical to those induced by live bacterial infection (130, 131). LPS-induced inflammation model presents well-known advantages, the primary one being that LPS does not replicate, allowing tight control of dosage and limiting the confounding nature of infection as compared to live bacteria models. LPS is commonly used to understand the complexities of the neuroimmune–neuroendocrine relationship and has been demonstrated to be a reliable activator of innate immune responses (97, 108) and HPA axis (66, 95, 108, 116, 132). Thus, LPS acts as an experimental systemic immunological stressor (133).

Lipopolysaccharide activates toll-like receptors and initiates a cascade of signalization leading to cytokine production that is crucial for infection clearance (134). Monocytes, neutrophils, macrophages, dendritic cells, and mast cells all express TLR4 at their surface membrane (135137). Upon activation of the TLR4/MD2 complex by LPS, a series of phosphorylation steps are activated, leading to the phosphorylation of inhibitory (I)κB, which releases nuclear factor (NF)-κB from its complex (138). NF-κB is subsequently translocated into the nucleus where it activates the transcription of pro-inflammatory cytokines such as IL-1β, TNFα, and IL-6, as well as anti-inflammatory cytokines such as IL-1 receptor antagonist (IL-1ra) and IL-10 (139, 140). Cytokines released in the blood stream are able to activate the release of cyclooxygenase (COX)-2 from the hypothalamus to induce hyperalgesia (141). COX-2 also stimulates the conversion of arachidonic acid into prostaglandins (PGE2), which acts in the vascular organ of the lamina terminalis and in the ventromedial preoptic area of the anterior hypothalamus to stimulate heat conservation via cutaneous vasoconstriction and attenuation of sweating, and heat production via increases in the metabolism of brown adipose tissue (142). Circulating IL-1β is also known to directly activate hypothalamic PVN to stimulate the release of corticosterone from adrenal cortex (143, 144). LPS activation of Kupffer cells in the liver is also known to activate the release of IL-1β that can contribute to hyperalgesia via vagal afferences (145), as vagotomy abolishes the LPS-induced hyperalgesia (145).

Neonatal LPS Exposure Changes Immune Responses Later in Life

Several lines of evidence from clinical and animal work suggest that exposure to LPS during the neonatal period is associated with altered immune responses later in life (66, 97, 109, 146150). Most importantly, long-term inflammatory responses within the CNS are greatly influenced by immunological stressors early in life. Incubation of cord blood from 1-month old children with LPS for 5 h resulted in increased mRNA expression of IL-6 and TNFα compared to cord blood from the same age incubated with medium (146). In rats, neonatal LPS exposure produces immediate upregulation of gene expression of chemokines and cytokines within the neonatal brain, as indicated by upregulation of mRNA levels of Ccl7, Cxcl1, Cxcl10, IL-1β, and IL-6 in the hippocampus 2 h following LPS exposure in rat pups at PND 4 (151). The effect of neonatal LPS exposure on cytokine levels in limbic areas can persist into adulthood. Our laboratory has previously shown that neonatal LPS exposure at PNDs 3 and 5 results in increased IL-1β and TNFα protein levels in the hippocampus following exposure to restraint stress in adulthood (66). Recent investigations point toward a critical role played by the hippocampus in modulating pain via upregulation of IL-1β expression (152). del Rey et al. documented a strong correlation between increased hippocampal IL-1β transcripts and mechanical allodynia in chronic constriction injury and spared nerve injury (SNI) models (152). However, it is not known whether changes in protein levels of IL-1β in the hippocampus contribute to increased pain sensitivity in inflammatory pain models (i.e., formalin test). Neonatal immune challenge has also been reported to alter febrile responses later in life (147, 148, 150). Fever is considered an important component of the innate immune response and is thought to play a crucial role in survival through its ability to efficiently clear the pathogen while limiting the extent of inflammatory damage (153, 154). Animals prevented from developing fever have higher risk of morbidity and mortality than animals that are allowed to develop fever (155). Rats exposed to LPS at P14 exhibited attenuated fever responses following a subsequent LPS challenge (147, 149) or stress (150) in adulthood. The effect of neonatal LPS exposure on adult febrile responses is thought to be mediated by pro-inflammatory cytokines, as neonatally LPS-treated rats displayed significantly reduced plasma levels of TNFα and IL-6 following subsequent LPS exposure in adulthood. This reduction in turn was strongly correlated with the observed attenuated febrile responses in LPS animals (147). Interestingly, basal maintenance of body temperature in adult rats was not affected by neonatal LPS administration (110). This finding implies that a single LPS exposure is not able to alter febrile responses later in life, but that a “second hit” is necessary to “unmask” the altered febrile responses following a neonatal immune challenge. Central levels of PGE2 and specifically in the preoptic region, a region involved in the febrigenic thermoeffector pathways (156, 157), have also been targeted as potential mechanisms mediating the attenuated febrile responses following a neonatal immune challenge. For instance, PGE2 levels in the preoptic area were increased in rats exposed to LPS at P14 (150). Additionally, glucocorticoids play a critical role in inducing the febrile response, as adrenalectomy or blockade of GRs using the GR antagonist RU-486 abolished the fever induced by neonatal exposure to LPS (147). Finally, our laboratory has previously demonstrated that rats exposed to LPS at PNDs 3 and 5 displayed increased susceptibility to tumor and lung metastases following exposure to stress in adulthood (97, 108). Moreover, neonatal immune challenge produced reduced NK cell activity and increased neuroendocrine responsivity to restraint stress in adulthood (97, 108).

Taken together, an early immunological stressor has profound effects on the immunological reaction pattern later in life, leading to altered neuroimmune function at subsequent exposures to immunological challenges. This implies that what the immune system of an organism has been exposed to very early in life will in fact define its capacity to defeat pathogens later in life.

Impact of Neonatal LPS Exposure on Endocrine Function

Microbial microbiota can affect the postnatal development of HPA axis, and an increasing body of evidence has demonstrated that neonatal exposure to LPS is associated with long-term alterations in HPA axis activity (66, 97, 116, 149, 158). Neonatal exposure to LPS during P3 and 5 has been reported to increase circulating levels of corticosterone at both time points (66, 132, 159), suggesting that neonatal LPS exposure is capable of altering HPA axis function during the SHRP. This alteration in HPA axis function following a neonatal immune challenge persists throughout the life of the animal. Adult rats treated with LPS as neonates displayed enhanced plasma corticosterone and ACTH levels in response to restraint stress, noise stress, or in response to a second LPS hit in adulthood (66, 95, 97, 116, 132). This altered peripheral endocrine response was also accompanied by central neuroendocrine changes, as indicated by increased CRH mRNA levels in the PVN and decreased GR density in the hypothalamus, hippocampus, and frontal cortex following exposure to stress in adulthood (95). These structures are known to mediate the inhibitory effects of glucocorticoids on CRH synthesis in the PVN and the release of ACTH following stress (160, 161), suggesting a decreased negative feedback sensitivity to glucocorticoids and, thus, an enhanced HPA responsiveness to stress following a neonatal immune challenge. We have demonstrated in our laboratory that dual exposure to LPS during P3 and P5 in rats is associated with increased circulating corticosterone at P7 and P22, but not P13, 1 h following injection of formalin into the hind paw (68). P22 rats neonatally treated with LPS also exhibited a trend toward decreased GR mRNA in the hypothalamus (68).

Overall, these data suggest that exposure to LPS during the neonatal period can reprogram the neuroendocrine axis. This reprogramming increases the reactivity of animals to a second physiological challenge later in life. Pain is an aversive experience and, therefore, capable of activating the HPA axis (162). Given that neonatal LPS exposure has been associated with increased release of peripheral and central pro-inflammatory cytokines later in life (66, 151) and considering the well-established role of pro-inflammatory cytokines in producing hyperalgesia (145), it is reasonable to assume that neonatal LPS exposure is likely to be associated with increased pain sensitivity later in life.

Impact of Neonatal Exposure to LPS on Nociceptive Responses

The first postnatal week (P7–P10) of rodent’s life is equivalent to the last trimester in humans (36–40 GW) in terms of brain growth, gliogenesis, axonal and dendritic density, as well as consolidation of the immune system (11, 163165). Preterm infants are, as discussed earlier, at high risk of infection during the neonatal period. Early-life infections in turn are known to be the cause of attenuated neurodevelopmental outcomes in these vulnerable infants (166). It is, therefore, important to address the impact of immune challenge on pain sensitivity later in life. Boisse et al found that administration of LPS at P14 in rats produced thermal and mechanical hyperalgesia that paralleled the enhanced expression of COX-2 protein levels in the lumbar spinal cord (141). Although this study did not directly demonstrate that the increased level of COX in the spinal cord contributed to the observed hyperalgesia in LPS-treated animals, it suggested a potential role of prostaglandins in mediating the LPS-induced hyperalgesia. Increased COX mRNA levels were also observed 4 h following LPS injection in P3 and P21 rats (P0 is birth) (167). A number of studies from our laboratory have indicated that dual exposure of LPS during P3 and 5 in rats produced long-term alterations in inflammatory pain responses later in life. Neonatal LPS administration evoked increased formalin-induced behavioral responses (i.e., flinching and licking) in P13, 22, and adult rats (4, 68, 168). The LPS-induced hyperalgesia observed in P22 rats coincided with altered HPA axis activity, as indicated by increased circulating corticosterone and decreased GR hypothalamic mRNA 1 h postformalin injection, as well as altered immune responses following formalin injection as indicated by increased mast cell degranulation and increased circulating IL-1β (4, 68). Moreover, the LPS-induced hyperalgesia in preadolescent rats was accompanied by altered spinal dorsal horn (SDH) intrinsic properties, as well as decreased neuronal activity (i.e., Fos expression) in the PAG (68, 168). LPS-treated adult rats exhibited hyperalgesia that coincided with central neuroimmune changes, as indicated by increased IL-1β in the hippocampus 1 h postformalin injection. No differences were observed in peripheral IL-1β release or mast cell degranulation (4). Although we reported enhanced hippocampal ILβ in LPS-treated adult rats, we do not know which immune cell releases this pro-inflammatory cytokine following neonatal immune challenge and subsequent inflammatory challenge. Of particular interest, hippocampal parenchyma astrocytes have been recently shown to produce the cytokine CCL2 24 h post-LPS injection in adult mice (169), suggesting an important role of astrocytes in the neuroinflammation produced by systemic LPS injection.

Taken together, these data challenge the traditional concept that pain is originating solely from activation of neurons and suggest that components of the immune system play an imminent role in modulating pain sensitivity. Using LPS as a model of infection, LPS-induced hyperalgesia arises by both peripheral and central mechanisms. Peripherally, LPS triggers, e.g., macrophages to release pro-inflammatory cytokines that sensitize nociceptors (145, 170, 171). In fact, LPS can directly activate TRPA1-expressing neurons independent of TLR4 (172). Centrally, LPS can activate microglial cells in the spinal cord and astrocytes in brain regions such as the hippocampus and produce hyperalgesia (169, 173).

The Neuroimmune Interface in Pain in the Adult Organism

As discussed so far, the exposure to immunological stressors very early in development of an individual has far-reaching effects on neural structure and function as well as on the immune and HPA axis activity. We have also pointed to defining changes for the adult pain system. In the mature body, the systems are fully developed and less malleable. However, the immune system continues to affect the function of the nervous system in a manner that drives pain sensitivity, by inducing functional changes. In this section, we describe some acute neuroimmune interactions in pain perception. Such neuroimmune interaction may potentially be of importance for the transition from acute to chronic pain in a long-term perspective.

Animal Studies Demonstrate Inflammation-Induced Pain Sensitivity

The role of the immune system was traditionally viewed as protecting the organism from invading pathogens. However, it is now well established that the bidirectional interaction between the immune and nervous systems plays a crucial role in pain modulation (125, 174177). Pro-inflammatory cytokines play an important role in this immune to brain bidirectional interaction (121, 145). When exposed to LPS, immune cells such as macrophages, monocytes, and mast cells release many pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6 into the circulation creating an “inflammatory soup” condition that enhances pain sensitivity by sensitizing nociceptors (178180). These pro-inflammatory cytokines also signal to the brain to induce a set of physiological responses including fever, lethargy, decreased social interaction, decreased sexual activity, and decreased food and water intake, increased circulating corticosterone, collectively known as sickness behavior (181183). Importantly, pain facilitation or hyperalgesia is considered to be an integral part of sickness behavior (121, 125). Peripheral inflammation can lead to central neuroinflammation via many different ways. First, through vagal afferences since subdiaphragmatic vagotomy reversed the hyperalgesia induced by IL-1β or LPS (145). Alternatively, cytokines can access the brain through areas that lack the blood–brain barrier (BBB) such as the organum vasculosum lamina terminalis (184). LPS produces IL-1β in the brain, which is initially restricted to choroid plexus and circumventricular organs, then diffuse to the brain side of BBB (185). Cytokines have also been suggested to enter the brain via active transport systems across the BBB (186, 187).

The first report on the impact of LPS exposure on pain responses was the study by Mason, who demonstrated that i.p. administration of LPS in adult rats significantly decreased tail flick latency, an effect that peaked at 1 h post-LPS administration (123). The LPS-induced thermal hyperalgesia observed in adult rats was reversed following the administration of IL-1ra (188), indicating that IL-1β is an important mediator of this hyperalgesia. Pro-inflammatory cytokines released by immune cells are known to induce hyperalgesia when administered both peripherally and centrally, particularly IL-1β (145, 170, 189, 190). For instance, intracerebroventricular (ICV) administration of the recombinant human IL-1β (rhIL-1β) in rats induced thermal hyperalgesia (170), while ICV injection of the IL-1β antagonist IL-1ra abolished this hyperalgesia (170). Intraplantar injection of IL-1β has been associated with increased discharge of SDH neurons in response to non-noxious stimuli (190). Local administration of IL-1ra decreased the LPS-induced hyperalgesia (171).

Interleukin-1β is also known to contribute to flinching responses in the formalin test given that an intraplantar injection in rats of antisera anti-IL-1β prior to formalin injection significantly attenuated flinching responses in the formalin test (191). We have previously shown that rats exposed to LPS during the neonatal period displayed increased circulating IL-1β at P22 in response to formalin injection (4). Adult rats previously subjected to neonatal immune challenge also displayed enhanced hippocampal IL-1β that coincides with the LPS-induced hyperalgesia at this age (4). The source of this hippocampal IL-1β is not known, but it is highly probable that it is originating from astrocytes or microglial cells within the hippocampus. Interestingly, at the same age (i.e., PND 22), and at the same time point following formalin injection (i.e., 1 h postformalin injection), we observed altered intrinsic properties of SDH, lamina I, and lamina II neurons in LPS-treated rats as indicated by lower input resistance compared to saline-treated rats (68).

Spinal dorsal horn neurons are the first component of the CNS to receive incoming noxious sensory information, and their output is determined by a combination of their synaptic inputs and intrinsic neuronal properties (192). Formalin injection is known to activate peripheral nerves, which results in turn in activation of dorsal horn neurons (193195). Hind paw injection of formalin is associated with the release of numerous substances in the spinal cord, including prostaglandin E2 (196). Bath application of prostaglandin E2 results in changes in intrinsic properties of dorsal horn neurons including decreased input resistance (197). Since this change was only observed in LPS-treated preadolescent rats, it is possible that the neonatal exposure to LPS resulted in either an increase in pro-inflammatory cytokines within the spinal cord or an increased susceptibility of SDH neurons to pro-inflammatory cytokines. This assumption is confirmed by the fact that intrathecal administration of IL-1ra has been reported to block formalin-induced hyperalgesia (198). The source of spinal hyperalgesia seems to involve microglia and astrocytes since intrathecal administration of fluorocitrate, an inhibitor of glial metabolic function, blocked the formalin-induced hyperalgesia (198).

Additionally, IL-1β has been documented to act supraspinally to induce hyperalgesia. For instance, microinjection of IL-1β into the preoptic area of the hypothalamus is sufficient to induce thermal hyperalgesia (199). Of particular interest is the observation that IP or ICV administration of IL-1β has been documented to produce an increase in plasma levels of corticosterone and ACTH, an action that is mediated by the release of CRH from the PVN (144, 200). The neonatal immune challenge is likely to influence the generation of new neurons in the hippocampus. This assumption is confirmed by the fact that an intraplantar injection of the nociceptive inflammatory agent Complete Freund’s Adjuvant at P8 results in more BrdU and doublecortin-labeled cells, both measures of newborn neurons, in the SGZ of the dentate gyrus (201). Whether such neurons release IL-1β in response to neonatal LPS exposure remains to be determined.

At the peripheral level, the enhanced IL-1β plasma levels observed at PND 22 in LPS-treated rats coincide with higher degree of mast cell degranulation, which was also accompanied by increased formalin-induced nociception (4). Mast cells are located in the vicinity of primary nociceptive neurons and vasculature and their degranulation has been reported to regulate the excitability of nociceptive nerve endings (202). Mast cell degranulation can also produce thermal hyperalgesia via the production of nerve growth factor (203). Previous studies have documented an important role of mast cells in formalin-induced nociception. Blocking mast cell activity using the mast cell stabilizer cromolyn abolished formalin-induced pain responses in the late phase (204). Interestingly, mast cells are also known to express receptor for IL-1β and to produce IL-1β following inflammation (205).

Inflammation-Induced Pain Sensitivity in Humans

The human physiology is much more sensitive to LPS provocation than that of rodents. To avoid the risk of sepsis, very low doses of LPS are used in humans (usually 0.2–4.0 ng/kg), the highest doses often requiring additional antipyretic pharmacological treatment. The most common dose for psychological research is around 0.4–1 ng/kg LPS from E. coli, which induces a clear rise of pro-inflammatory cytokines TNFα, IL-1β, IL-6, and IL-8 in the blood (206208). Human studies can also benefit from vaccinations of healthy individuals as an inflammatory model, and patients undergoing immunotherapy can be studied. The behavioral outcomes of experimental immune activation are very similar to sickness behavior exhibited by experimental animals; individuals report increased anxiety, worsened mood, and increased pain sensitivity (205, 209, 210). Appetite is reduced, and fatigue and anhedonia increase parallel to decreased social interest (126). The immune activation also disrupts memory and cognition and changes motivation (6, 211, 212). In human studies with the lowest LPS doses, the effects can in fact be so subtle that blinding can be maintained.

Pain Sensitivity during Immune Provocation

So far, only LPS stimulations have been used to study the pain system specifically in humans, and several studies have shown that experimental immune activation increases pain sensitivity in humans, too. Deep (muscular and visceral) pain is more readily affected than superficial (cutaneous and mechanical) pain (207, 213, 214). Also, the change in pain sensitivity usually correlates with peripheral cytokine levels. As in all experimental pain research, the mode of pain stimulation as well as the pain intensity applied may affect the outcome. Threshold pain is not processed exactly the same way as suprathreshold (intense) pain, and pain from within the body is relayed to the brain in pathways partly distinct from those used to relay cutaneous pain (215). Also, the nociceptive effect may depend on the immunological pressure, i.e., the LPS dose in experimental models. Two studies show that threshold pressure pain sensitivity is affected the same way in men and women, despite the generally higher cytokine levels found in women during LPS stimulation (207, 216). Interestingly, no sex differences in psychological outcomes, such as anxiety or perceived health, are seen either despite the sex differences in cytokine release (207, 208). One study has, however, shown that women are indeed more affected by inflammation with regard to pain perception (207). In this study, the descending pain inhibition of women was weakened during LPS stimulation, while men remained unaffected. In parallel, women were more pain sensitive to intense cutaneous pain, too, while men only changed their perception of deep pain. Furthermore, one study using a high LPS dose (2.0 ng/kg) has in fact shown increased pain sensitivity to intense cutaneous pain in men. Sex differences in inflammation-induced pain sensitivity need further exploration. An intriguing mechanism for a potential sex difference was recently suggested in a murine study (217), where female mice did not require microglia activation to develop pain hypersensitivity, but appeared to have alternative routes via the adaptive immune system. This alternative route did not seem accessible to males. Future research will have to establish if these mechanisms are relevant for humans as well and their role in immune-driven pain sensitivity. Furthermore, sex-dependent alterations in neuroendocrine function in human subjects following LPS provocation have been shown (218). Healthy humans exhibited enhanced circulating levels of cortisol (peak response at 5 h post-LPS injection) after LPS injection (208, 219). The effect appears to be more pronounced in women (208), but the data are inconclusive (219). On a final note, experimental pain is sensitive to stress, which could potentially be a confounder in LPS studies on pain. Perhaps, surprisingly, however, stress levels generally remain low among the participants throughout the studies (207). Our experience is that because LPS stimulations, due to ethical considerations using bacterial endotoxin injections in healthy subjects, require very clear participant information and a hospital environment with experienced personnel and constant supervision, participants describe a feeling of safety and control even at higher, quite uncomfortable doses (such as 2.0 ng/kg).

Brain Activity during Experimental Immune Activation

Although the cytokines released during immune activation may affect and sensitize peripheral nerve endings, the main effect by which the immune system changes the function of the nervous system during sickness is believed to occur centrally via induced sickness behavior. It is reasonable to assume that changes in the emotional circuitries underlying the increased anxiety and depressed mood seen during immune activation may also lead to increased pain sensitivity due to overlapping function with the medial (affective) pain network (215), such as the amygdala, the cingulate, and prefrontal cortices. Also, as sickness is per definition an interoceptive signal, i.e., a signal of the internal state of the body (220), areas involved in interoception and homeostasis such as the insular cortex, which is also part of the pain network, could potentially be affected. Several studies have attempted to elucidate the neural correlates of sickness behavior in the human brain. Most studies have used functional magnetic resonance imaging (fMRI) with cognitive and emotional paradigms. The main methodological limitation for this type of research is the fact that only the lower LPS doses used in humans are compatible with a brain scanning protocol, i.e., those that do not induce nausea or shivering.

Only two studies have explored pain perception directly during brain imaging so far, one using visceral pain stimuli (deep pain measurement) and mechanical pinprick pain (cutaneous pain measurement) (221) and the other using pressure pain (deep pain) (222). Benson et al. (221) showed increased activation within the posterior insula, dorsolateral PFC, anterior midcingulate, and somatosensory cortices for visceral pain stimulation, but not mechanical pain provocation. These areas are involved in pain and affective processing, interoception, and homeostatic regulation. Karshikoff et al. (222) described decreased activity after LPS injection in the lateral PFC and rostral anterior cingulate cortex (ACC), areas involved in descending pain inhibition, which may point to an increase in inflammation-induced pain sensitivity via diminished endogenous pain regulation. Additionally, the LPS group showed increased pain-dependent activity in the anterior insular cortex compared to placebo.

Emotional and cognitive fMRI paradigms corroborate the involvement of the cingulate, insula, and prefrontal cortices when the brain adapts to immune activation (221, 223227), which are core areas in affective pain processing and pain regulation. Using a vaccination protocol as experimental immune provocation, Harrison et al. have shown increased activity in the subgenual ACC during emotional stimuli and in areas involved in interoceptive function during a Stoop task, such as the brain stem, the cingulate, and anterior insula (225, 226). To maintain the same level of performance during peripheral inflammatory activity, regions of the PFC appear to be required (224, 225)—areas implicated in pain regulation and processing of affective components of pain. In several studies, the increased BOLD activity in these areas correlates with peripheral cytokine levels (210, 222, 226, 228, 229).

Immune challenge affects the levels of neurotransmitters in the brain (6, 230). The expression of sickness behavior can potentially be manipulated by drugs affecting neurotransmitter levels such as serotonin reuptake inhibitors, which are compounds often used to ameliorate chronic pain. Hannestad et al. (231) have, for example, shown that the effects on fatigue are ameliorated by pretreatment of serotonin reuptake inhibitors, but not by dopamine and noradrenaline reuptake inhibitor. Peripherally induced inflammation has also been shown to activate microglia directly (232, 233). This is of special importance for chronic pain, as microglia have been implicated in the establishment of chronic pain (121).

In the past decade, it has thus been shown that acute inflammation induces pain sensitivity in humans as well. Most importantly, acute inflammation has a global effect on brain function, modulating the neural function in several brain areas involved in pain perception. Although the experimental models used are of an acute character, similar mechanisms are likely to be involved when the organism is subdued to long-term inflammatory activity.

The HPA System and Pain in Adult Organisms

Pain is not only modulated by immunological stressors but also by activation of the HPA axis. Pain is a sensory as well as an emotional experience. It is by nature a stressful event and, therefore, capable of activating the HPA axis. As we have mentioned, there is a large individual variability in developing chronic pain. One possible mechanism that may account for this individual variability in pain responses is how each individual responds to stressful events. Exaggeration or maladaptive response following stress may lead to altered pain responses. The HPA axis involves a defined neural circuit that comprises many brain regions including the amygdala, the mPFC, and the hippocampus. These areas are also important in pain modulation (234237). In other words, a non-painful stressful stimulus is able to recruit parts of the same neural network involved in the pain response. Therefore, under conditions of stress, pain sensitivity may be exaggerated. Indeed, activation of CRH receptors in the amygdala facilitated pain responses through increased excitatory postsynaptic current in the parabrachio-amygdaloid synapse in rodents (238). Furthermore, administration of CRH into the CeA increased visceral nociception, as indicated by exaggerated number of abdominal muscle contractions in response to colorectal distension (239). On the other hand, the contribution of acute stress in analgesia commonly known as “stress-induced analgesia” has been traditionally well documented (240, 241), and at this point in time the exact contribution of cortisol in modulating pain is still a matter of debate within the scientific community.

In human clinical samples, some researchers have found that low back pain and enhanced musculoskeletal pain are often associated with hypocortisolemia (242, 243), while others demonstrated that patients suffering from chronic back pain displayed higher levels of cortisol compared to control group (244). This hypercortisolemia was associated with smaller hippocampal volume and higher pain-evoked response in the anterior parahippocampal gyrus (244). This variability in cortisolemia in pain condition not only may be due to the intensity of the stress response (245) but may also well depend on the neural circuit recruited following the stress stimulus, as the neural circuits within PVN are quite complex, and the final outcome depends on the nature of the stressor [for review, please see Ref. (237)]. In inflammatory pain model, such as the formalin test in rodents, LPS-induced hyperalgesia in infant and preadolescent rats coincided with increased circulating corticosterone 1 h following intraplantar injection of formalin (68). However, a recent study demonstrated that elevated levels of plasma corticosterone produced analgesia via attenuated C fiber-mediated spinal responses (246).

Overall, the abovementioned animal and human studies suggest that changes in HPA axis activity can contribute to pain. Although more studies are needed to confirm the exact contribution of cortisol (in humans) or corticosterone (in rodents) in modulating pain responses, the involvement of neuroendocrine response in pain is evident. Therefore, new therapeutic approaches, which not only target neural activity but also the neuroendocrine axis, are needed to treat chronic pain patients.

A Lifetime Perspective

Although the acute effects of immune provocation on pain sensitivity are fairly well documented by now, as described in the previous sections, long-term inflammatory effects are not well understood. At this point in time, the most research on long-term effects of inflammatory activity on behavior has focused on depression. In humans, one incentive to study the mechanisms of sickness behavior came from clinical observations of immunotherapy eliciting side effects that resemble sickness behavior, such as depressive symptoms, fatigue, and aches. In, for example, hepatitis C patients undergoing IFN-α therapy, up to 45% of the patients develop depression (247). The typical signs of sickness behavior appear at the commencement of immunotherapy, whereas the establishment of depression requires time, and potentially, persistent inflammatory input during this time. It is now argued that depression is in part an inflammatory disease (248), and that a subgroup of clinically depressed patients suffers from a chronic low-grade systemic inflammation. Childhood trauma has also been shown to predispose persons to depression, but potentially not only via learning and HPA dysregulation as traditionally suggested but also via inflammation. Depressed patients with a history of traumatic events have higher low-grade inflammatory activity (249). Most interestingly, these patients benefit from pharmacological treatments that combine anti-inflammatory compounds and traditional antidepressants (249). Suggested mechanisms between inflammatory activity and depression include cytokines, serotonin, HPA dysregulation, GABA, and glutamate, all of which are neuroimmune pathways also implicated in pain [for extensive reviews see, e.g., Ref. (6, 250)]. Recent research is now shifting the focus toward similar mechanisms for chronic pain and fatigue (6, 230).

Inflammatory Disease and Pain

Chronic pain is a common comorbid symptom to many inflammatory diseases (251). Moreover, coronary heart disease (252), metabolic disorders (253), and life stress (254) increase the risk of developing chronic pain. It has been suggested that one of the underlying mechanisms for this association is indeed inflammation (252254). Furthermore, chronic pain has been associated with low-grade inflammation (255). Mechanistically, peripheral chronic inflammation may become chronic within the CNS via changes in the central immune responses, by means of mechanism previously discussed. In animals, transient peripheral infections and inflammations or chronic exposure to low level (subclinical) inflammations can either activate microglia directly (256, 257) or “prime” the cells so that a recurrent inflammatory provocation becomes more severe (258). A systemic inflammatory challenge leads to an exaggerated fever response and sickness behavior in the presence of “primed” microglia in rodents (259, 260). “Priming” of immune components, or the requirement of a “second immunological hit” to reveal susceptibility as discussed previously, is exemplified in a recent clinical study. Obesity has been associated with chronic pain and is considered a chronic low-grade inflammatory state (253). Obesity did not predict postsurgical pain intensity or inflammatory levels (255). BMI did, however, correlate with the increased immune response of leukocytes after LPS stimulation, suggesting sensitivity to inflammatory development in the obese patients that could results in complications associated with inflammation further down the road, such as chronic pain. Another study points to differences in pharmacological treatment strategies on pain after surgery, depending on prior inflammatory disease. Non-steroidal anti-inflammatory drugs had a better protective effect against the development of long-term pain after surgery in patients with a background of inflammatory disease, than opioids (261).

The Immune System Develops throughout Life

As discussed previously, the immune system carries the imprint of early-life inflammatory events. However, the function of immune system in fighting previously unencountered pathogens and protect the organism on reinfection relies on the ability to adapt and learn throughout life. Immune functioning is determined partly by genetics (262) and varies greatly between individuals. Individuals differ in their susceptibility to different types of infections, such as bacterial, viral, and fungal (262), and several single nucleotide polymorphisms related to immune pathways have been described (262). For example, the IL-6 and IL-8 pathways appear to have large genetic variations between individuals, while the IL-1 pathway has remained more conserved throughout evolution (262). Recent research emphasizes the importance of experience in shaping the adult immune response, similar to what has been described for infants in the previous sections. In fact, most of the individual differences seen in immune function in adult humans stem from non-heritable changes (263, 264). The immune system activates distinct cytokine patterns depending on the type of infection, and continuously learns from experience to adapt its inflammatory response (265). In theory, each person thus possesses an immune system that is a product of the types, strengths, and number of infections, diseases, and injuries encountered throughout life. Prior experience should thus impact future immunological reaction patterns. Epidemiological studies on comorbidity and risk factors for common disease give support to the idea that lifetime immune challenges affect disease susceptibility. A recent study shows that in patients with multimorbidity (in this specific study more than 10 disease diagnoses), the incidence of lifetime infections, inflammation, injuries, and tumors was 7–10 times as common as in a primary health care population (266). Lifetime accumulation of strong immune activation may thus potentially lead to increased general disease susceptibility and comorbidity (266). Furthermore, lifetime inflammatory disease is a risk factor for developing neurodegenerative disease (267269). A plausible mechanism is that the accumulation of inflammatory activity in the body induces neuroinflammation in the brain, which in turn affects the function of the CNS (267).

When Adaptation Becomes a Liability

The process of perinatal programming posits that exposure to environmental factors during a sensitive window of development is able to program or have long-term consequences on physiological systems later in life. A fundamental aspect of perinatal programming is that developing organisms “sense” the early-life environment and use this information to establish homeostatic set points (270, 271). This process of perinatal programming has evolved as an adaptive mechanism enabling the fetus to constantly interact with the maternal environment (via the placenta) and use this information as a forecast of the environmental conditions it will eventually face postnatally. As such, preparing it to adjust its physiological and behavioral need to match the requirements of the ex utero world (272). In this perspective, fetal programming is an example of predictive adaptive responses where the fetus uses present cues to shape an adaptive phenotype to future environmental stimuli (31, 273). However, this adjustment can become maladaptive in the case where a “mismatch” exists between the expected ex utero environment and the actual circumstances. More importantly, when adverse events occur during a critical window of vulnerability of physiological systems that are still undergoing fine-tuning and plasticity, an individual may become predisposed to high susceptibility and exaggerated sensitivity to environmental stimuli later in life.

Correspondingly, the immune system and the HPA system adapt and change according to the stressors that the individual encounters throughout life, in order to maintain health and homeostasis. Pain is one of the most important survival signals available to us, and a life without pain perception is often a short one, as can be seen in individuals with congenital insensitivity to pain (274). However, when the imprint of the different stressors throughout life accumulate, interact, and/or become prolonged, the consequence may be detrimental for the pain system. For diseases like chronic pain, with such wide individual variability in symptomatology and treatment efficacy (8, 9), not only should comorbid disease and stressful life events (i.e., concurrent with the pain) be considered when exploring the pathophysiology but also past stressors. In this study, we want to increase the awareness of the profound effect of the immune system on the pain system from birth to old age, via neuroimmune and neuroendocrine interactions. In other words, the faith of the pain system starts in utero.

Conclusion

In this review, we argue that the individual differences in the susceptibility to chronic pain and success of treatment thereof may be the result of the person’s prenatal history, combined with childhood as well as lifetime experience. We have highlighted the biological underpinnings and potential consequences on the pain system induced by the stress and infectious/inflammatory load an individual is subjected to. The neuroimmune and neuroendocrine interactions that affect the pain system start in the womb and modulate the pain system throughout life. The modulations may be of both structural and functional nature and may be both adaptive and maladaptive. In order to understand individual differences in pain, human studies of long-term effects of inflammatory stressors are needed.

Author Contributions

IZ and BK wrote the manuscript and approved the final version.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The reviewer AF-H declared a shared affiliation, though no other collaboration, with the authors to the handling Editor, who ensured that the process nevertheless met the standards of a fair and objective review.

Funding

IZ is an overseas researcher under Postdoctoral Fellowship of Japan Society for the Promotion of Science (JSPS). BK is supported by AFA Insurance and the Swedish Society of Medicine.

References

1. Chapman CR, Tuckett RP, Song CW. Pain and stress in a systems perspective: reciprocal neural, endocrine, and immune interactions. J Pain (2008) 9:122–45. doi:10.1016/j.jpain.2007.09.006

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Walker SM, Beggs S, Baccei ML. Persistent changes in peripheral and spinal nociceptive processing after early tissue injury. Exp Neurol (2016) 275(Pt 2):253–60. doi:10.1016/j.expneurol.2015.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Loram LC, Taylor FR, Strand KA, Frank MG, Sholar P, Harrison JA, et al. Prior exposure to glucocorticoids potentiates lipopolysaccharide induced mechanical allodynia and spinal neuroinflammation. Brain Behav Immun (2011) 25:1408–15. doi:10.1016/j.bbi.2011.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Zouikr I, Ahmed AF, Horvat JC, Beagley KW, Clifton VL, Ray A, et al. Programming of formalin-induced nociception by neonatal LPS exposure: maintenance by peripheral and central neuroimmune activity. Brain Behav Immun (2015) 44:235–46. doi:10.1016/j.bbi.2014.10.014

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Hains LE, Loram LC, Weiseler JL, Frank MG, Bloss EB, Sholar P, et al. Pain intensity and duration can be enhanced by prior challenge: initial evidence suggestive of a role of microglial priming. J Pain (2010) 11:1004–14. doi:10.1016/j.jpain.2010.01.271

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Walker AK, Kavelaars A, Heijnen CJ, Dantzer R. Neuroinflammation and comorbidity of pain and depression. Pharmacol Rev (2014) 66:80–101. doi:10.1124/pr.113.008144

CrossRef Full Text | Google Scholar

7. Veehof MM, Trompetter HR, Bohlmeijer ET, Schreurs KM. Acceptance- and mindfulness-based interventions for the treatment of chronic pain: a meta-analytic review. Cogn Behav Ther (2016) 45:5–31. doi:10.1080/16506073.2015.1098724

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Ramond-Roquin A, Bouton C, Begue C, Petit A, Roquelaure Y, Huez JF. Psychosocial risk factors, interventions, and comorbidity in patients with non-specific low back pain in primary care: need for comprehensive and patient-centered care. Front Med (2015) 2:73. doi:10.3389/fmed.2015.00073

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Hechler T, Kanstrup M, Holley AL, Simons LE, Wicksell R, Hirschfeld G, et al. Systematic review on intensive interdisciplinary pain treatment of children with chronic pain. Pediatrics (2015) 136:115–27. doi:10.1542/peds.2014-3319

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Hensch TK. Critical period regulation. Annu Rev Neurosci (2004) 27:549–79. doi:10.1146/annurev.neuro.27.070203.144327

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Cowan WM. The development of the brain. Sci Am (1979) 241:113–33. doi:10.1038/scientificamerican0979-112

CrossRef Full Text | Google Scholar

12. Kostovic I, Judas M, Rados M, Hrabac P. Laminar organization of the human fetal cerebrum revealed by histochemical markers and magnetic resonance imaging. Cereb Cortex (2002) 12:536–44. doi:10.1093/cercor/12.5.536

CrossRef Full Text | Google Scholar

13. Jones LS, Gauger LL, Davis JN, Slotkin TA, Bartolome JV. Postnatal development of brain alpha 1-adrenergic receptors: in vitro autoradiography with [125I]HEAT in normal rats and rats treated with alpha-difluoromethylornithine, a specific, irreversible inhibitor of ornithine decarboxylase. Neuroscience (1985) 15:1195–202. doi:10.1016/0306-4522(85)90262-3

CrossRef Full Text | Google Scholar

14. Huttenlocher PR, Dabholkar AS. Regional differences in synaptogenesis in human cerebral cortex. J Comp Neurol (1997) 387:167–78. doi:10.1002/(SICI)1096-9861(19971020)387:2<167::AID-CNE1>3.0.CO;2-Z

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Bourgeois JP. Synaptogenesis, heterochrony and epigenesis in the mammalian neocortex. Acta Paediatr Suppl (1997) 422:27–33. doi:10.1111/j.1651-2227.1997.tb18340.x

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Bluml S, Wisnowski JL, Nelson MD Jr, Paquette L, Gilles FH, Kinney HC, et al. Metabolic maturation of the human brain from birth through adolescence: insights from in vivo magnetic resonance spectroscopy. Cereb Cortex (2013) 23:2944–55. doi:10.1093/cercor/bhs283

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Giedd JN, Lalonde FM, Celano MJ, White SL, Wallace GL, Lee NR, et al. Anatomical brain magnetic resonance imaging of typically developing children and adolescents. J Am Acad Child Adolesc Psychiatry (2009) 48:465–70. doi:10.1097/CHI.0b013e31819f2715

CrossRef Full Text | Google Scholar

18. Dehaene-Lambertz G, Spelke ES. The infancy of the human brain. Neuron (2015) 88:93–109. doi:10.1016/j.neuron.2015.09.026

CrossRef Full Text | Google Scholar

19. Alkonyi B, Juhasz C, Muzik O, Behen ME, Jeong JW, Chugani HT. Thalamocortical connectivity in healthy children: asymmetries and robust developmental changes between ages 8 and 17 years. AJNR Am J Neuroradiol (2011) 32:962–9. doi:10.3174/ajnr.A2417

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Hubel DH, Wiesel TN. The period of susceptibility to the physiological effects of unilateral eye closure in kittens. J Physiol (1970) 206:419–36. doi:10.1113/jphysiol.1970.sp009022

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Gould E, Reeves AJ, Graziano MS, Gross CG. Neurogenesis in the neocortex of adult primates. Science (1999) 286:548–52. doi:10.1126/science.286.5439.548

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Kaplan MS, Hinds JW. Neurogenesis in the adult rat: electron microscopic analysis of light radioautographs. Science (1977) 197:1092–4. doi:10.1126/science.887941

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Gould E, Vail N, Wagers M, Gross CG. Adult-generated hippocampal and neocortical neurons in macaques have a transient existence. Proc Natl Acad Sci U S A (2001) 98:10910–7. doi:10.1073/pnas.181354698

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Inta D, Alfonso J, von Engelhardt J, Kreuzberg MM, Meyer AH, van Hooft JA, et al. Neurogenesis and widespread forebrain migration of distinct GABAergic neurons from the postnatal subventricular zone. Proc Natl Acad Sci U S A (2008) 105:20994–9. doi:10.1073/pnas.0807059105

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Altman J. Autoradiographic and histological studies of postnatal neurogenesis. IV. Cell proliferation and migration in the anterior forebrain, with special reference to persisting neurogenesis in the olfactory bulb. J Comp Neurol (1969) 137:433–57. doi:10.1002/cne.901370404

CrossRef Full Text | Google Scholar

26. Merzenich MM, Nelson RJ, Stryker MP, Cynader MS, Schoppmann A, Zook JM. Somatosensory cortical map changes following digit amputation in adult monkeys. J Comp Neurol (1984) 224:591–605. doi:10.1002/cne.902240408

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Robertson D, Irvine DR. Plasticity of frequency organization in auditory cortex of guinea pigs with partial unilateral deafness. J Comp Neurol (1989) 282:456–71. doi:10.1002/cne.902820311

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Yang Y, Cheng Z, Tang H, Jiao H, Sun X, Cui Q, et al. Neonatal maternal separation impairs prefrontal cortical myelination and cognitive functions in rats through activation of Wnt signaling. Cereb Cortex (2016) 1–14. doi:10.1093/cercor/bhw121

CrossRef Full Text | Google Scholar

29. Metz AE, Yau HJ, Centeno MV, Apkarian AV, Martina M. Morphological and functional reorganization of rat medial prefrontal cortex in neuropathic pain. Proc Natl Acad Sci U S A (2009) 106:2423–8. doi:10.1073/pnas.0809897106

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Soztutar E, Colak E, Ulupinar E. Gender- and anxiety level-dependent effects of perinatal stress exposure on medial prefrontal cortex. Exp Neurol (2016) 275(Pt 2):274–84. doi:10.1016/j.expneurol.2015.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Gluckman PD, Hanson MA. Living with the past: evolution, development, and patterns of disease. Science (2004) 305:1733–6. doi:10.1126/science.1095292

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Gluckman PD, Hanson MA, Bateson P, Beedle AS, Law CM, Bhutta ZA, et al. Towards a new developmental synthesis: adaptive developmental plasticity and human disease. Lancet (2009) 373:1654–7. doi:10.1016/S0140-6736(09)60234-8

CrossRef Full Text | Google Scholar

33. Buss C, Entringer S, Wadhwa PD. Fetal programming of brain development: intrauterine stress and susceptibility to psychopathology. Sci Signal (2012) 5:t7. doi:10.1126/scisignal.2003406

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Mirnics K, Koerber HR. Prenatal development of rat primary afferent fibers: II. Central projections. J Comp Neurol (1995) 355:601–14. doi:10.1002/cne.903550409

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Beggs S, Torsney C, Drew LJ, Fitzgerald M. The postnatal reorganization of primary afferent input and dorsal horn cell receptive fields in the rat spinal cord is an activity-dependent process. Eur J Neurosci (2002) 16:1249–58. doi:10.1046/j.1460-9568.2002.02185.x

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Beggs S, Alvares D, Moss A, Currie G, Middleton J, Salter MW, et al. A role for NT-3 in the hyperinnervation of neonatally wounded skin. Pain (2012) 153:2133–9. doi:10.1016/j.pain.2012.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Reynolds ML, Fitzgerald M. Long-term sensory hyperinnervation following neonatal skin wounds. J Comp Neurol (1995) 358:487–98. doi:10.1002/cne.903580403

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Zouikr I, Tadros MA, Clifton VL, Beagley KW, Hodgson DM. Low formalin concentrations induce fine-tuned responses that are sex and age-dependent: a developmental study. PLoS One (2013) 8:e53384. doi:10.1371/journal.pone.0053384

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Teng CJ, Abbott FV. The formalin test: a dose-response analysis at three developmental stages. Pain (1998) 76:337–47. doi:10.1016/S0304-3959(98)00065-7

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Hathway GJ, Koch S, Low L, Fitzgerald M. The changing balance of brainstem-spinal cord modulation of pain processing over the first weeks of rat postnatal life. J Physiol (2009) 587:2927–35. doi:10.1113/jphysiol.2008.168013

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Kwok CH, Devonshire IM, Bennett AJ, Hathway GJ. Postnatal maturation of endogenous opioid systems within the periaqueductal grey and spinal dorsal horn of the rat. Pain (2014) 155:168–78. doi:10.1016/j.pain.2013.09.022

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Hathway GJ, Vega-Avelaira D, Fitzgerald M. A critical period in the supraspinal control of pain: opioid-dependent changes in brainstem rostroventral medulla function in preadolescence. Pain (2012) 153:775–83. doi:10.1016/j.pain.2011.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Zouikr I, Bartholomeusz MD, Hodgson DM. Early life programming of pain: focus on neuroimmune to endocrine communication. J Transl Med (2016) 14:123. doi:10.1186/s12967-016-0879-8

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Seckl JR. Glucocorticoids, developmental ‘programming’ and the risk of affective dysfunction. Prog Brain Res (2008) 167:17–34. doi:10.1016/S0079-6123(07)67002-2

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Matthews SG. Antenatal glucocorticoids and programming of the developing CNS. Pediatr Res (2000) 47:291–300. doi:10.1203/00006450-200003000-00003

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Groeneweg FL, Karst H, de Kloet ER, Joels M. Mineralocorticoid and glucocorticoid receptors at the neuronal membrane, regulators of nongenomic corticosteroid signalling. Mol Cell Endocrinol (2012) 350:299–309. doi:10.1016/j.mce.2011.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Riley SC, Challis JR. Corticotrophin-releasing hormone production by the placenta and fetal membranes. Placenta (1991) 12:105–19. doi:10.1016/0143-4004(91)90015-8

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Mastorakos G, Ilias I. Maternal and fetal hypothalamic-pituitary-adrenal axes during pregnancy and postpartum. Ann N Y Acad Sci (2003) 997:136–49. doi:10.1196/annals.1290.016

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Welberg LA, Seckl JR, Holmes MC. Inhibition of 11beta-hydroxysteroid dehydrogenase, the foeto-placental barrier to maternal glucocorticoids, permanently programs amygdala GR mRNA expression and anxiety-like behaviour in the offspring. Eur J Neurosci (2000) 12:1047–54. doi:10.1046/j.1460-9568.2000.00958.x

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Ohkawa T, Takeshita S, Murase T, Kambegawa A, Okinaga S, Arai K. Ontogeny of the response of the hypothalamo-pituitary-adrenal axis to maternal immobilization stress in rats. Endocrinol Jpn (1991) 38:187–94. doi:10.1507/endocrj1954.38.187

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Aldenhoff JB, Gruol DL, Rivier J, Vale W, Siggins GR. Corticotropin releasing factor decreases postburst hyperpolarizations and excites hippocampal neurons. Science (1983) 221:875–7. doi:10.1126/science.6603658

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Noorlander CW, De Graan PN, Middeldorp J, Van Beers JJ, Visser GH. Ontogeny of hippocampal corticosteroid receptors: effects of antenatal glucocorticoids in human and mouse. J Comp Neurol (2006) 499:924–32. doi:10.1002/cne.21162

CrossRef Full Text | Google Scholar

53. Bresson JL, Clavequin MC, Fellmann D, Bugnon C. Anatomical and ontogenetic studies of the human paraventriculo-infundibular corticoliberin system. Neuroscience (1985) 14:1077–90. doi:10.1016/0306-4522(85)90278-7

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Thliveris JA, Currie RW. Observations on the hypothalamo-hypophyseal portal vasculature in the developing human fetus. Am J Anat (1980) 157:441–4. doi:10.1002/aja.1001570411

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Brosnan PG. The hypothalamic pituitary axis in the fetus and newborn. Semin Perinatol (2001) 25:371–84. doi:10.1053/sper.2001.29038

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Asa SL, Kovacs K, Laszlo FA, Domokos I, Ezrin C. Human fetal adenohypophysis. Histologic and immunocytochemical analysis. Neuroendocrinology (1986) 43:308–16. doi:10.1159/000124545

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Diaz R, Brown RW, Seckl JR. Distinct ontogeny of glucocorticoid and mineralocorticoid receptor and 11beta-hydroxysteroid dehydrogenase types I and II mRNAs in the fetal rat brain suggest a complex control of glucocorticoid actions. J Neurosci (1998) 18:2570–80.

PubMed Abstract | Google Scholar

58. Yi SJ, Masters JN, Baram TZ. Glucocorticoid receptor mRNA ontogeny in the fetal and postnatal rat forebrain. Mol Cell Neurosci (1994) 5:385–93. doi:10.1006/mcne.1994.1048

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Levine S. The ontogeny of the hypothalamic-pituitary-adrenal axis. The influence of maternal factors. Ann N Y Acad Sci (1994) 746:275–88; discussion 289–93. doi:10.1111/j.1749-6632.1994.tb39245.x

CrossRef Full Text | Google Scholar

60. Stanton ME, Wallstrom J, Levine S. Maternal contact inhibits pituitary-adrenal stress responses in preweanling rats. Dev Psychobiol (1987) 20:131–45. doi:10.1002/dev.420200204

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Schmidt MV, Enthoven L, van der Mark M, Levine S, de Kloet ER, Oitzl MS. The postnatal development of the hypothalamic-pituitary-adrenal axis in the mouse. Int J Dev Neurosci (2003) 21:125–32. doi:10.1016/S0736-5748(03)00030-3

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Pryce CR, Feldon J, Fuchs E, Knuesel I, Oertle T, Sengstag C, et al. Postnatal ontogeny of hippocampal expression of the mineralocorticoid and glucocorticoid receptors in the common marmoset monkey. Eur J Neurosci (2005) 21:1521–35. doi:10.1111/j.1460-9568.2005.04003.x

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Hagberg H, Bona E, Gilland E, Puka-Sundvall M. Hypoxia-ischaemia model in the 7-day-old rat: possibilities and shortcomings. Acta Paediatr Suppl (1997) 422:85–8. doi:10.1111/j.1651-2227.1997.tb18353.x

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Dobbing J. The Later Development of the Brain and Its Vulnerability. London: Heinemann (1981). p. 744–58.

Google Scholar

65. Anisman H, Zaharia MD, Meaney MJ, Merali Z. Do early-life events permanently alter behavioral and hormonal responses to stressors? Int J Dev Neurosci (1998) 16:149–64. doi:10.1016/S0736-5748(98)00025-2

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Walker AK, Nakamura T, Hodgson DM. Neonatal lipopolysaccharide exposure alters central cytokine responses to stress in adulthood in Wistar rats. Stress (2010) 13:506–15. doi:10.3109/10253890.2010.489977

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Nakamura T, Walker AK, Sominsky L, Allen T, Rosengren S, Hodgson DM. Maternal separation in early life impairs tumor immunity in adulthood in the F344 rat. Stress (2011) 14:335–43. doi:10.3109/10253890.2010.548014

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Zouikr I, Tadros MA, Barouei J, Beagley KW, Clifton VL, Callister RJ, et al. Altered nociceptive, endocrine, and dorsal horn neuron responses in rats following a neonatal immune challenge. Psychoneuroendocrinology (2014) 41:1–12. doi:10.1016/j.psyneuen.2013.11.016

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Shanks N, Meaney MJ. Hypothalamic-pituitary-adrenal activation following endotoxin administration in the developing rat: a CRH-mediated effect. J Neuroendocrinol (1994) 6:375–83. doi:10.1111/j.1365-2826.1994.tb00596.x

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Hemming VG, Overall JC Jr, Britt MR. Nosocomial infections in a newborn intensive-care unit. Results of forty-one months of surveillance. N Engl J Med (1976) 294:1310–6. doi:10.1056/NEJM197606102942403

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Scheible KM, Emo J, Yang H, Holden-Wiltse J, Straw A, Huyck H, et al. Developmentally determined reduction in CD31 during gestation is associated with CD8+ T cell effector differentiation in preterm infants. Clin Immunol (2015) 161:65–74. doi:10.1016/j.clim.2015.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Fadel S, Sarzotti M. Cellular immune responses in neonates. Int Rev Immunol (2000) 19:173–93. doi:10.3109/08830180009088504

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Taylor S, Bryson YJ. Impaired production of gamma-interferon by newborn cells in vitro is due to a functionally immature macrophage. J Immunol (1985) 134:1493–7.

PubMed Abstract | Google Scholar

74. Trivedi HN, HayGlass KT, Gangur V, Allardice JG, Embree JE, Plummer FA. Analysis of neonatal T cell and antigen presenting cell functions. Hum Immunol (1997) 57:69–79. doi:10.1016/S0198-8859(97)00202-4

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Gentile LF, Nacionales DC, Lopez MC, Vanzant E, Cuenca A, Cuenca AG, et al. Protective immunity and defects in the neonatal and elderly immune response to sepsis. J Immunol (2014) 192:3156–65. doi:10.4049/jimmunol.1301726

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Kollmann TR, Crabtree J, Rein-Weston A, Blimkie D, Thommai F, Wang XY, et al. Neonatal innate TLR-mediated responses are distinct from those of adults. J Immunol (2009) 183:7150–60. doi:10.4049/jimmunol.0901481

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Adkins B, Hamilton K. Freshly isolated, murine neonatal T cells produce IL-4 in response to anti-CD3 stimulation. J Immunol (1992) 149:3448–55.

PubMed Abstract | Google Scholar

78. Garcia AM, Fadel SA, Cao S, Sarzotti M. T cell immunity in neonates. Immunol Res (2000) 22:177–90. doi:10.1385/IR:22:2-3:177

CrossRef Full Text | Google Scholar

79. Lee SM, Suen Y, Qian J, Knoppel E, Cairo MS. The regulation and biological activity of interleukin 12. Leuk Lymphoma (1998) 29:427–38. doi:10.3109/10428199809050903

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Urban JF Jr, Fayer R, Chen SJ, Gause WC, Gately MK, Finkelman FD. IL-12 protects immunocompetent and immunodeficient neonatal mice against infection with Cryptosporidium parvum. J Immunol (1996) 156:263–8.

PubMed Abstract | Google Scholar

81. Arulanandam BP, Van Cleave VH, Metzger DW. IL-12 is a potent neonatal vaccine adjuvant. Eur J Immunol (1999) 29:256–64. doi:10.1002/(SICI)1521-4141(199901)29:01<256::AID-IMMU256>3.0.CO;2-G

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Olson JA, McDonald-Hyman C, Jameson SC, Hamilton SE. Effector-like CD8(+) T cells in the memory population mediate potent protective immunity. Immunity (2013) 38:1250–60. doi:10.1016/j.immuni.2013.05.009

CrossRef Full Text | Google Scholar

83. Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature (2016) 532(7600):512–6. doi:10.1038/nature17655

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med (2001) 29:1303–10. doi:10.1097/00003246-200107000-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Rennie J. Robertson’s Textbook of Neonatology. London: Churchill Livingstone (2005).

Google Scholar

86. Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol (2004) 4:553–64. doi:10.1038/nri1394

CrossRef Full Text | Google Scholar

87. Levy O. Innate immunity of the human newborn: distinct cytokine responses to LPS and other Toll-like receptor agonists. J Endotoxin Res (2005) 11:113–6. doi:10.1179/096805105X37376

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Levy O. Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol (2007) 7:379–90. doi:10.1038/nri2075

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Levy O, Zarember KA, Roy RM, Cywes C, Godowski PJ, Wessels MR. Selective impairment of TLR-mediated innate immunity in human newborns: neonatal blood plasma reduces monocyte TNF-alpha induction by bacterial lipopeptides, lipopolysaccharide, and imiquimod, but preserves the response to R-848. J Immunol (2004) 173:4627–34. doi:10.4049/jimmunol.173.7.4627

PubMed Abstract | CrossRef Full Text | Google Scholar

90. De Wit D, Tonon S, Olislagers V, Goriely S, Boutriaux M, Goldman M, et al. Impaired responses to toll-like receptor 4 and toll-like receptor 3 ligands in human cord blood. J Autoimmun (2003) 21:277–81. doi:10.1016/j.jaut.2003.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Koenig JM, Yoder MC. Neonatal neutrophils: the good, the bad, and the ugly. Clin Perinatol (2004) 31:39–51. doi:10.1016/j.clp.2004.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Osrin D, Vergnano S, Costello A. Serious bacterial infections in newborn infants in developing countries. Curr Opin Infect Dis (2004) 17:217–24. doi:10.1097/00001432-200406000-00008

CrossRef Full Text | Google Scholar

93. Skogstrand K, Hougaard DM, Schendel DE, Bent NP, Svaerke C, Thorsen P. Association of preterm birth with sustained postnatal inflammatory response. Obstet Gynecol (2008) 111:1118–28. doi:10.1097/AOG.0b013e31817057fb

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Marshall-Clarke S, Reen D, Tasker L, Hassan J. Neonatal immunity: how well has it grown up? Immunol Today (2000) 21:35–41. doi:10.1016/S0167-5699(99)01548-0

CrossRef Full Text | Google Scholar

95. Shanks N, Larocque S, Meaney MJ. Neonatal endotoxin exposure alters the development of the hypothalamic-pituitary-adrenal axis: early illness and later responsivity to stress. J Neurosci (1995) 15:376–84.

Google Scholar

96. Cai Z, Pan ZL, Pang Y, Evans OB, Rhodes PG. Cytokine induction in fetal rat brains and brain injury in neonatal rats after maternal lipopolysaccharide administration. Pediatr Res (2000) 47:64–72. doi:10.1203/00006450-200001000-00013

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Hodgson DM, Knott B, Walker FR. Neonatal endotoxin exposure influences HPA responsivity and impairs tumor immunity in Fischer 344 rats in adulthood. Pediatr Res (2001) 50:750–5. doi:10.1203/00006450-200112000-00020

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Meyer U, Feldon J, Schedlowski M, Yee BK. Immunological stress at the maternal-foetal interface: a link between neurodevelopment and adult psychopathology. Brain Behav Immun (2006) 20:378–88. doi:10.1016/j.bbi.2005.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Pang Y, Cai Z, Rhodes PG. Disturbance of oligodendrocyte development, hypomyelination and white matter injury in the neonatal rat brain after intracerebral injection of lipopolysaccharide. Brain Res Dev Brain Res (2003) 140:205–14. doi:10.1016/S0165-3806(02)00606-5

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Urakubo A, Jarskog LF, Lieberman JA, Gilmore JH. Prenatal exposure to maternal infection alters cytokine expression in the placenta, amniotic fluid, and fetal brain. Schizophr Res (2001) 47:27–36. doi:10.1016/S0920-9964(00)00032-3

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Byrne M, Agerbo E, Bennedsen B, Eaton WW, Mortensen PB. Obstetric conditions and risk of first admission with schizophrenia: a Danish national register based study. Schizophr Res (2007) 97:51–9. doi:10.1016/j.schres.2007.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Brown AS, Cohen P, Harkavy-Friedman J, Babulas V, Malaspina D, Gorman JM, et al. A.E. Bennett research award. Prenatal rubella, premorbid abnormalities, and adult schizophrenia. Biol Psychiatry (2001) 49:473–86. doi:10.1016/S0006-3223(01)01068-X

CrossRef Full Text | Google Scholar

103. Buka SL, Cannon TD, Torrey EF, Yolken RH. Maternal exposure to herpes simplex virus and risk of psychosis among adult offspring. Biol Psychiatry (2008) 63:809–15. doi:10.1016/j.biopsych.2007.09.022

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Brown AS, Susser ES. In utero infection and adult schizophrenia. Ment Retard Dev Disabil Res Rev (2002) 8:51–7. doi:10.1002/mrdd.10004

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Fortier ME, Joober R, Luheshi GN, Boksa P. Maternal exposure to bacterial endotoxin during pregnancy enhances amphetamine-induced locomotion and startle responses in adult rat offspring. J Psychiatr Res (2004) 38:335–45. doi:10.1016/j.jpsychires.2003.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Yolken RH, Karlsson H, Yee F, Johnston-Wilson NL, Torrey EF. Endogenous retroviruses and schizophrenia. Brain Res Brain Res Rev (2000) 31:193–9. doi:10.1016/S0165-0173(99)00037-5

CrossRef Full Text | Google Scholar

107. Kwan ML, Metayer C, Crouse V, Buffler PA. Maternal illness and drug/medication use during the period surrounding pregnancy and risk of childhood leukemia among offspring. Am J Epidemiol (2007) 165:27–35. doi:10.1093/aje/kwj336

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Hodgson DM, Knott B. Potentiation of tumor metastasis in adulthood by neonatal endotoxin exposure: sex differences. Psychoneuroendocrinology (2002) 27:791–804. doi:10.1016/S0306-4530(01)00080-4

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Spencer SJ, Martin S, Mouihate A, Pittman QJ. Early-life immune challenge: defining a critical window for effects on adult responses to immune challenge. Neuropsychopharmacology (2006) 31:1910–8. doi:10.1038/sj.npp.1301004

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Spencer SJ, Field E, Pittman QJ. Neonatal programming by neuroimmune challenge: effects on responses and tolerance to septic doses of lipopolysaccharide in adult male and female rats. J Neuroendocrinol (2010) 22:272–81. doi:10.1111/j.1365-2826.2010.01967.x

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Walker FR, Hodyl NA, Krivanek KM, Hodgson DM. Early life host-bacteria relations and development: long-term individual differences in neuroimmune function following neonatal endotoxin challenge. Physiol Behav (2006) 87:126–34. doi:10.1016/j.physbeh.2005.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Walker FR, Owens J, Ali S, Hodgson DM. Individual differences in glucose homeostasis: do our early life interactions with bacteria matter? Brain Behav Immun (2006) 20:401–9. doi:10.1016/j.bbi.2005.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Wigle DT, Arbuckle TE, Turner MC, Berube A, Yang Q, Liu S, et al. Epidemiologic evidence of relationships between reproductive and child health outcomes and environmental chemical contaminants. J Toxicol Environ Health B Crit Rev (2008) 11:373–517. doi:10.1080/10937400801921320

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Sominsky L, Meehan CL, Walker AK, Bobrovskaya L, McLaughlin EA, Hodgson DM. Neonatal immune challenge alters reproductive development in the female rat. Horm Behav (2012) 62:345–55. doi:10.1016/j.yhbeh.2012.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Kajantie E. Fetal origins of stress-related adult disease. Ann N Y Acad Sci (2006) 1083:11–27. doi:10.1196/annals.1367.026

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, et al. Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proc Natl Acad Sci U S A (2000) 97:5645–50. doi:10.1073/pnas.090571897

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Sun Y, Vestergaard M, Christensen J, Olsen J. Prenatal exposure to elevated maternal body temperature and risk of epilepsy in childhood: a population-based pregnancy cohort study. Paediatr Perinat Epidemiol (2011) 25:53–9. doi:10.1111/j.1365-3016.2010.01143.x

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Rasmussen SA, Jamieson DJ, Bresee JS. Pandemic influenza and pregnant women. Emerg Infect Dis (2008) 14:95–100. doi:10.3201/eid1401.070667

CrossRef Full Text | Google Scholar

119. Meyer U, Feldon J, Yee BK. A review of the fetal brain cytokine imbalance hypothesis of schizophrenia. Schizophr Bull (2009) 35:959–72. doi:10.1093/schbul/sbn022

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Dombrowski SC, Martin RP, Huttunen MO. Association between maternal fever and psychological/behavior outcomes: a hypothesis. Birth Defects Res A Clin Mol Teratol (2003) 67:905–10. doi:10.1002/bdra.10096

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Watkins LR, Maier SF, Goehler LE. Immune activation: the role of pro-inflammatory cytokines in inflammation, illness responses and pathological pain states. Pain (1995) 63:289–302. doi:10.1016/0304-3959(95)00186-7

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Hutchinson MR, Buijs M, Tuke J, Kwok YH, Gentgall M, Williams D, et al. Low-dose endotoxin potentiates capsaicin-induced pain in man: evidence for a pain neuroimmune connection. Brain Behav Immun (2013) 30:3–11. doi:10.1016/j.bbi.2013.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Mason P. Lipopolysaccharide induces fever and decreases tail flick latency in awake rats. Neurosci Lett (1993) 154:134–6. doi:10.1016/0304-3940(93)90189-R

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Watkins LR, Maier SF. Immune regulation of central nervous system functions: from sickness responses to pathological pain. J Intern Med (2005) 257:139–55. doi:10.1111/j.1365-2796.2004.01443.x

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Watkins LR, Maier SF. The pain of being sick: implications of immune-to-brain communication for understanding pain. Annu Rev Psychol (2000) 51:29–57. doi:10.1146/annurev.psych.51.1.29

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun (2007) 21:153–60. doi:10.1016/j.bbi.2006.09.006

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Yirmiya R, Pollak Y, Morag M, Reichenberg A, Barak O, Avitsur R, et al. Illness, cytokines, and depression. Ann N Y Acad Sci (2000) 917:478–87. doi:10.1111/j.1749-6632.2000.tb05412.x

CrossRef Full Text | Google Scholar

128. Pearce BD. Schizophrenia and viral infection during neurodevelopment: a focus on mechanisms. Mol Psychiatry (2001) 6:634–46. doi:10.1038/sj.mp.4000956

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Yoon BH, Romero R, Kim CJ, Jun JK, Gomez R, Choi JH, et al. Amniotic fluid interleukin-6: a sensitive test for antenatal diagnosis of acute inflammatory lesions of preterm placenta and prediction of perinatal morbidity. Am J Obstet Gynecol (1995) 172:960–70. doi:10.1016/0002-9378(95)90028-4

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Burrell R. Human responses to bacterial endotoxin. Circ Shock (1994) 43:137–53.

PubMed Abstract | Google Scholar

131. Rosenberger CM, Scott MG, Gold MR, Hancock RE, Finlay BB. Salmonella typhimurium infection and lipopolysaccharide stimulation induce similar changes in macrophage gene expression. J Immunol (2000) 164:5894–904. doi:10.4049/jimmunol.164.11.5894

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Walker AK, Nakamura T, Byrne RJ, Naicker S, Tynan RJ, Hunter M, et al. Neonatal lipopolysaccharide and adult stress exposure predisposes rats to anxiety-like behaviour and blunted corticosterone responses: implications for the double-hit hypothesis. Psychoneuroendocrinology (2009) 34:1515–25. doi:10.1016/j.psyneuen.2009.05.010

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo-pituitary-adrenal (HPA) axis. J Endotoxin Res (2003) 9:3–24. doi:10.1177/09680519030090010101

CrossRef Full Text | Google Scholar

134. Raetz CR, Whitfield C. Lipopolysaccharide endotoxins. Annu Rev Biochem (2002) 71:635–700. doi:10.1146/annurev.biochem.71.110601.135414

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Diks SH, van Deventer SJ, Peppelenbosch MP. Lipopolysaccharide recognition, internalisation, signalling and other cellular effects. J Endotoxin Res (2001) 7:335–48. doi:10.1177/09680519010070050101

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Applequist SE, Wallin RP, Ljunggren HG. Variable expression of toll-like receptor in murine innate and adaptive immune cell lines. Int Immunol (2002) 14:1065–74. doi:10.1093/intimm/dxf069

PubMed Abstract | CrossRef Full Text | Google Scholar

137. McCurdy JD, Lin TJ, Marshall JS. Toll-like receptor 4-mediated activation of murine mast cells. J Leukoc Biol (2001) 70:977–84.

PubMed Abstract | Google Scholar

138. Doyle SL, O’Neill LA. Toll-like receptors: from the discovery of NFkappaB to new insights into transcriptional regulations in innate immunity. Biochem Pharmacol (2006) 72:1102–13. doi:10.1016/j.bcp.2006.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Cartmell T, Ball C, Bristow AF, Mitchell D, Poole S. Endogenous interleukin-10 is required for the defervescence of fever evoked by local lipopolysaccharide-induced and Staphylococcus aureus-induced inflammation in rats. J Physiol (2003) 549:653–64. doi:10.1113/jphysiol.2002.037291

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Conti B, Tabarean I, Andrei C, Bartfai T. Cytokines and fever. Front Biosci (2004) 9:1433–49. doi:10.2741/1341

CrossRef Full Text | Google Scholar

141. Boisse L, Spencer SJ, Mouihate A, Vergnolle N, Pittman QJ. Neonatal immune challenge alters nociception in the adult rat. Pain (2005) 119:133–41. doi:10.1016/j.pain.2005.09.022

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Morrison SF, Nakamura K, Madden CJ. Central control of thermogenesis in mammals. Exp Physiol (2008) 93:773–97. doi:10.1113/expphysiol.2007.041848

CrossRef Full Text | Google Scholar

143. Besedovsky H, del Rey A, Sorkin E, Dinarello CA. Immunoregulatory feedback between interleukin-1 and glucocorticoid hormones. Science (1986) 233:652–4. doi:10.1126/science.3014662

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Berkenbosch F, van Oers J, del Rey A, Tilders F, Besedovsky H. Corticotropin-releasing factor-producing neurons in the rat activated by interleukin-1. Science (1987) 238:524–6. doi:10.1126/science.2443979

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF. Characterization of cytokine-induced hyperalgesia. Brain Res (1994) 654:15–26. doi:10.1016/0006-8993(94)91566-0

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Belderbos ME, van Bleek GM, Levy O, Blanken MO, Houben ML, Schuijff L, et al. Skewed pattern of toll-like receptor 4-mediated cytokine production in human neonatal blood: low LPS-induced IL-12p70 and high IL-10 persist throughout the first month of life. Clin Immunol (2009) 133:228–37. doi:10.1016/j.clim.2009.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Ellis S, Mouihate A, Pittman QJ. Early life immune challenge alters innate immune responses to lipopolysaccharide: implications for host defense as adults. FASEB J (2005) 19:1519–21. doi:10.1096/fj.04-3569fje

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Ellis S, Mouihate A, Pittman QJ. Neonatal programming of the rat neuroimmune response: stimulus specific changes elicited by bacterial and viral mimetics. J Physiol (2006) 571:695–701. doi:10.1113/jphysiol.2005.102939

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Boisse L, Mouihate A, Ellis S, Pittman QJ. Long-term alterations in neuroimmune responses after neonatal exposure to lipopolysaccharide. J Neurosci (2004) 24:4928–34. doi:10.1523/JNEUROSCI.1077-04.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Soriano RN, Branco LG. Reduced stress fever is accompanied by increased glucocorticoids and reduced PGE2 in adult rats exposed to endotoxin as neonates. J Neuroimmunol (2010) 225:77–81. doi:10.1016/j.jneuroim.2010.04.018

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Schwarz JM, Bilbo SD. LPS elicits a much larger and broader inflammatory response than Escherichia coli infection within the hippocampus of neonatal rats. Neurosci Lett (2011) 497:110–5. doi:10.1016/j.neulet.2011.04.042

PubMed Abstract | CrossRef Full Text | Google Scholar

152. del Rey A, Yau HJ, Randolf A, Centeno MV, Wildmann J, Martina M, et al. Chronic neuropathic pain-like behavior correlates with IL-1beta expression and disrupts cytokine interactions in the hippocampus. Pain (2011) 152:2827–35. doi:10.1016/j.pain.2011.09.013

CrossRef Full Text | Google Scholar

153. Schobitz B, Reul JM, Holsboer F. The role of the hypothalamic-pituitary-adrenocortical system during inflammatory conditions. Crit Rev Neurobiol (1994) 8:263–91.

PubMed Abstract | Google Scholar

154. Jiang Q, DeTolla L, van Rooijen N, Singh IS, Fitzgerald B, Lipsky MM, et al. Febrile-range temperature modifies early systemic tumor necrosis factor alpha expression in mice challenged with bacterial endotoxin. Infect Immun (1999) 67:1539–46.

Google Scholar

155. Kluger MJ, Kozak W, Conn CA, Leon LR, Soszynski D. Role of fever in disease. Ann N Y Acad Sci (1998) 856:224–33. doi:10.1111/j.1749-6632.1998.tb08329.x

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Feleder C, Perlik V, Blatteis CM. Preoptic norepinephrine mediates the febrile response of guinea pigs to lipopolysaccharide. Am J Physiol Regul Integr Comp Physiol (2007) 293:R1135–43. doi:10.1152/ajpregu.00067.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Scammell TE, Elmquist JK, Griffin JD, Saper CB. Ventromedial preoptic prostaglandin E2 activates fever-producing autonomic pathways. J Neurosci (1996) 16:6246–54.

PubMed Abstract | Google Scholar

158. Iwasa T, Matsuzaki T, Kinouchi R, Fujisawa S, Murakami M, Kiyokawa M, et al. Neonatal LPS injection alters the body weight regulation systems of rats under non-stress and immune stress conditions. Int J Dev Neurosci (2010) 28:119–24. doi:10.1016/j.ijdevneu.2009.08.015

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Walker FR, Brogan A, Smith R, Hodgson DM. A profile of the immediate endocrine, metabolic and behavioural responses following a dual exposure to endotoxin in early life. Physiol Behav (2004) 83:495–504. doi:10.1016/j.physbeh.2004.08.030

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Sapolsky RM, Armanini MP, Packan DR, Sutton SW, Plotsky PM. Glucocorticoid feedback inhibition of adrenocorticotropic hormone secretagogue release. Relationship to corticosteroid receptor occupancy in various limbic sites. Neuroendocrinology (1990) 51:328–36. doi:10.1159/000125357

CrossRef Full Text | Google Scholar

161. Kovacs K, Kiss JZ, Makara GB. Glucocorticoid implants around the hypothalamic paraventricular nucleus prevent the increase of corticotropin-releasing factor and arginine vasopressin immunostaining induced by adrenalectomy. Neuroendocrinology (1986) 44:229–34. doi:10.1159/000124650

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Blackburn-Munro G. Hypothalamo-pituitary-adrenal axis dysfunction as a contributory factor to chronic pain and depression. Curr Pain Headache Rep (2004) 8:116–24. doi:10.1007/s11916-004-0025-9

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Dobbing J, Sands J. Comparative aspects of the brain growth spurt. Early Hum Dev (1979) 3:79–83. doi:10.1016/0378-3782(79)90022-7

CrossRef Full Text | Google Scholar

164. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci (2009) 32:149–84. doi:10.1146/annurev.neuro.051508.135600

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Holsapple MP, West LJ, Landreth KS. Species comparison of anatomical and functional immune system development. Birth Defects Res B Dev Reprod Toxicol (2003) 68:321–34. doi:10.1002/bdrb.10035

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz SR, Vohr B, et al. Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA (2004) 292:2357–65. doi:10.1001/jama.292.19.2357

CrossRef Full Text | Google Scholar

167. Hunter D, Chai C, Barr GA. Effects of COX inhibition and LPS on formalin induced pain in the infant rat. Dev Neurobiol (2015) 75:1068–79. doi:10.1002/dneu.22230

PubMed Abstract | CrossRef Full Text | Google Scholar

168. Zouikr I, James MH, Campbell EJ, Clifton VL, Beagley KW, Dayas CV, et al. Altered formalin-induced pain and Fos induction in the periaqueductal grey of preadolescent rats following neonatal LPS exposure. PLoS One (2014) 9:e98382. doi:10.1371/journal.pone.0098382

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Hasegawa-Ishii S, Inaba M, Umegaki H, Unno K, Wakabayashi K, Shimada A. Endotoxemia-induced cytokine-mediated responses of hippocampal astrocytes transmitted by cells of the brain-immune interface. Sci Rep (2016) 6:25457. doi:10.1038/srep25457

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Oka T, Aou S, Hori T. Intracerebroventricular injection of interleukin-1 beta induces hyperalgesia in rats. Brain Res (1993) 624:61–8. doi:10.1016/0006-8993(93)90060-Z

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Cunha JM, Cunha FQ, Poole S, Ferreira SH. Cytokine-mediated inflammatory hyperalgesia limited by interleukin-1 receptor antagonist. Br J Pharmacol (2000) 130:1418–24. doi:10.1038/sj.bjp.0703434

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Meseguer V, Alpizar YA, Luis E, Tajada S, Denlinger B, Fajardo O, et al. TRPA1 channels mediate acute neurogenic inflammation and pain produced by bacterial endotoxins. Nat Commun (2014) 5:3125. doi:10.1038/ncomms4125

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Yoon SY, Patel D, Dougherty PM. Minocycline blocks lipopolysaccharide induced hyperalgesia by suppression of microglia but not astrocytes. Neuroscience (2012) 221:214–24. doi:10.1016/j.neuroscience.2012.06.024

PubMed Abstract | CrossRef Full Text | Google Scholar

174. Grace PM, Hutchinson MR, Maier SF, Watkins LR. Pathological pain and the neuroimmune interface. Nat Rev Immunol (2014) 14:217–31. doi:10.1038/nri3621

CrossRef Full Text | Google Scholar

175. Watkins LR, Maier SF. Implications of immune-to-brain communication for sickness and pain. Proc Natl Acad Sci U S A (1999) 96:7710–3. doi:10.1073/pnas.96.14.7710

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Ren K, Dubner R. Interactions between the immune and nervous systems in pain. Nat Med (2010) 16:1267–76. doi:10.1038/nm.2234

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci (2005) 6:521–32. doi:10.1038/nrn1700

CrossRef Full Text | Google Scholar

178. DeLeo JA, Colburn RW, Nichols M, Malhotra A. Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J Interferon Cytokine Res (1996) 16:695–700. doi:10.1089/jir.1996.16.695

CrossRef Full Text | Google Scholar

179. Reeve AJ, Patel S, Fox A, Walker K, Urban L. Intrathecally administered endotoxin or cytokines produce allodynia, hyperalgesia and changes in spinal cord neuronal responses to nociceptive stimuli in the rat. Eur J Pain (2000) 4:247–57. doi:10.1053/eujp.2000.0177

PubMed Abstract | CrossRef Full Text | Google Scholar

180. Sung CS, Wen ZH, Chang WK, Ho ST, Tsai SK, Chang YC, et al. Intrathecal interleukin-1beta administration induces thermal hyperalgesia by activating inducible nitric oxide synthase expression in the rat spinal cord. Brain Res (2004) 1015:145–53. doi:10.1016/j.brainres.2004.04.068

PubMed Abstract | CrossRef Full Text | Google Scholar

181. Dantzer R. Cytokine-induced sickness behaviour: a neuroimmune response to activation of innate immunity. Eur J Pharmacol (2004) 500:399–411. doi:10.1016/j.ejphar.2004.07.040

PubMed Abstract | CrossRef Full Text | Google Scholar

182. Dantzer R. Cytokine-induced sickness behavior: where do we stand? Brain Behav Immun (2001) 15:7–24. doi:10.1006/brbi.2000.0613

PubMed Abstract | CrossRef Full Text | Google Scholar

183. Konsman JP, Parnet P, Dantzer R. Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci (2002) 25:154–9. doi:10.1016/S0166-2236(00)02088-9

PubMed Abstract | CrossRef Full Text | Google Scholar

184. Katsuura G, Arimura A, Koves K, Gottschall PE. Involvement of organum vasculosum of lamina terminalis and preoptic area in interleukin 1 beta-induced ACTH release. Am J Physiol (1990) 258:E163–71.

PubMed Abstract | Google Scholar

185. Konsman JP, Kelley K, Dantzer R. Temporal and spatial relationships between lipopolysaccharide-induced expression of Fos, interleukin-1beta and inducible nitric oxide synthase in rat brain. Neuroscience (1999) 89:535–48. doi:10.1016/S0306-4522(98)00368-6

PubMed Abstract | CrossRef Full Text | Google Scholar

186. Banks WA, Kastin AJ, Ehrensing CA. Blood-borne interleukin-1 alpha is transported across the endothelial blood-spinal cord barrier of mice. J Physiol (1994) 479(Pt 2):257–64. doi:10.1113/jphysiol.1994.sp020293

CrossRef Full Text | Google Scholar

187. Banks WA, Niehoff ML, Zalcman SS. Permeability of the mouse blood-brain barrier to murine interleukin-2: predominance of a saturable efflux system. Brain Behav Immun (2004) 18:434–42. doi:10.1016/j.bbi.2003.09.013

PubMed Abstract | CrossRef Full Text | Google Scholar

188. Maier SF, Wiertelak EP, Martin D, Watkins LR. Interleukin-1 mediates the behavioral hyperalgesia produced by lithium chloride and endotoxin. Brain Res (1993) 623:321–4. doi:10.1016/0006-8993(93)91446-Y

PubMed Abstract | CrossRef Full Text | Google Scholar

189. Ferreira SH, Lorenzetti BB, Bristow AF, Poole S. Interleukin-1 beta as a potent hyperalgesic agent antagonized by a tripeptide analogue. Nature (1988) 334:698–700. doi:10.1038/334698a0

PubMed Abstract | CrossRef Full Text | Google Scholar

190. Fukuoka H, Kawatani M, Hisamitsu T, Takeshige C. Cutaneous hyperalgesia induced by peripheral injection of interleukin-1 beta in the rat. Brain Res (1994) 657:133–40. doi:10.1016/0006-8993(94)90960-1

CrossRef Full Text | Google Scholar

191. Granados-Soto V, Alonso-Lopez R, Asomoza-Espinosa R, Rufino MO, Gomes-Lopes LD, Ferreira SH. Participation of COX, IL-1 beta and TNF alpha in formalin-induced inflammatory pain. Proc West Pharmacol Soc (2001) 44:15–7.

Google Scholar

192. Hille B. Ionic Channels of Excitable Membranes. Sunderland, MA: Sinauer Associates, Inc. (2001).

Google Scholar

193. Dickenson AH, Sullivan AF. Subcutaneous formalin-induced activity of dorsal horn neurones in the rat: differential response to an intrathecal opiate administered pre or post formalin. Pain (1987) 30:349–60. doi:10.1016/0304-3959(87)90023-6

PubMed Abstract | CrossRef Full Text | Google Scholar

194. Dickenson AH, Sullivan AF. Peripheral origins and central modulation of subcutaneous formalin-induced activity of rat dorsal horn neurones. Neurosci Lett (1987) 83:207–11. doi:10.1016/0304-3940(87)90242-4

PubMed Abstract | CrossRef Full Text | Google Scholar

195. Puig S, Sorkin LS. Formalin-evoked activity in identified primary afferent fibers: systemic lidocaine suppresses phase-2 activity. Pain (1996) 64:345–55. doi:10.1016/0304-3959(95)00121-2

PubMed Abstract | CrossRef Full Text | Google Scholar

196. Malmberg AB, Yaksh TL. The effect of morphine on formalin-evoked behaviour and spinal release of excitatory amino acids and prostaglandin E2 using microdialysis in conscious rats. Br J Pharmacol (1995) 114:1069–75. doi:10.1111/j.1476-5381.1995.tb13315.x

PubMed Abstract | CrossRef Full Text | Google Scholar

197. Baba H, Kohno T, Moore KA, Woolf CJ. Direct activation of rat spinal dorsal horn neurons by prostaglandin E2. J Neurosci (2001) 21:1750–6.

PubMed Abstract | Google Scholar

198. Watkins LR, Martin D, Ulrich P, Tracey KJ, Maier SF. Evidence for the involvement of spinal cord glia in subcutaneous formalin induced hyperalgesia in the rat. Pain (1997) 71:225–35. doi:10.1016/S0304-3959(97)03369-1

PubMed Abstract | CrossRef Full Text | Google Scholar

199. Oka T, Oka K, Hosoi M, Aou S, Hori T. The opposing effects of interleukin-1 beta microinjected into the preoptic hypothalamus and the ventromedial hypothalamus on nociceptive behavior in rats. Brain Res (1995) 700:271–8. doi:10.1016/0006-8993(95)00980-5

PubMed Abstract | CrossRef Full Text | Google Scholar

200. Sapolsky R, Rivier C, Yamamoto G, Plotsky P, Vale W. Interleukin-1 stimulates the secretion of hypothalamic corticotropin-releasing factor. Science (1987) 238:522–4. doi:10.1126/science.2821621

PubMed Abstract | CrossRef Full Text | Google Scholar

201. Leslie AT, Akers KG, Martinez-Canabal A, Mello LE, Covolan L, Guinsburg R. Neonatal inflammatory pain increases hippocampal neurogenesis in rat pups. Neurosci Lett (2011) 501:78–82. doi:10.1016/j.neulet.2011.06.047

PubMed Abstract | CrossRef Full Text | Google Scholar

202. Kovacs P, Hernadi I, Wilhelm M. Mast cells modulate maintained neuronal activity in the thalamus in vivo. J Neuroimmunol (2006) 171:1–7. doi:10.1016/j.jneuroim.2005.07.026

CrossRef Full Text | Google Scholar

203. Lewin GR, Rueff A, Mendell LM. Peripheral and central mechanisms of NGF-induced hyperalgesia. Eur J Neurosci (1994) 6:1903–12. doi:10.1111/j.1460-9568.1994.tb00581.x

PubMed Abstract | CrossRef Full Text | Google Scholar

204. Parada CA, Tambeli CH, Cunha FQ, Ferreira SH. The major role of peripheral release of histamine and 5-hydroxytryptamine in formalin-induced nociception. Neuroscience (2001) 102:937–44. doi:10.1016/S0306-4522(00)00523-6

PubMed Abstract | CrossRef Full Text | Google Scholar

205. Silver R, Curley JP. Mast cells on the mind: new insights and opportunities. Trends Neurosci (2013) 36:513–21. doi:10.1016/j.tins.2013.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

206. Martich GD, Boujoukos AJ, Suffredini AF. Response of man to endotoxin. Immunobiology (1993) 187:403–16. doi:10.1016/S0171-2985(11)80353-0

PubMed Abstract | CrossRef Full Text | Google Scholar

207. Karshikoff B, Lekander M, Soop A, Lindstedt F, Ingvar M, Kosek E, et al. Modality and sex differences in pain sensitivity during human endotoxemia. Brain Behav Immun (2015) 46:35–43. doi:10.1016/j.bbi.2014.11.014

PubMed Abstract | CrossRef Full Text | Google Scholar

208. Engler H, Benson S, Wegner A, Spreitzer I, Schedlowski M, Elsenbruch S. Men and women differ in inflammatory and neuroendocrine responses to endotoxin but not in the severity of sickness symptoms. Brain Behav Immun (2016) 52:18–26. doi:10.1016/j.bbi.2015.08.013

CrossRef Full Text | Google Scholar

209. Lasselin J, Elsenbruch S, Lekander M, Axelsson J, Karshikoff B, Grigoleit JS, et al. Mood disturbance during experimental endotoxemia: predictors of state anxiety as a psychological component of sickness behavior. Brain Behav Immun (2016) 57:30–7. doi:10.1016/j.bbi.2016.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

210. Inagaki TK, Muscatell KA, Irwin MR, Moieni M, Dutcher JM, Jevtic I, et al. The role of the ventral striatum in inflammatory-induced approach toward support figures. Brain Behav Immun (2015) 44:247–52. doi:10.1016/j.bbi.2014.10.006

CrossRef Full Text | Google Scholar

211. Harrison NA, Voon V, Cercignani M, Cooper EA, Pessiglione M, Critchley HD. A neurocomputational account of how inflammation enhances sensitivity to punishments versus rewards. Biol Psychiatry (2016) 80:73–81. doi:10.1016/j.biopsych.2015.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

212. Krabbe KS, Reichenberg A, Yirmiya R, Smed A, Pedersen BK, Bruunsgaard H. Low-dose endotoxemia and human neuropsychological functions. Brain Behav Immun (2005) 19:453–60. doi:10.1016/j.bbi.2005.04.010

PubMed Abstract | CrossRef Full Text | Google Scholar

213. de Goeij M, van Eijk LT, Vanelderen P, Wilder-Smith OH, Vissers KC, van der Hoeven JG, et al. Systemic inflammation decreases pain threshold in humans in vivo. PLoS One (2013) 8:e84159. doi:10.1371/journal.pone.0084159

PubMed Abstract | CrossRef Full Text | Google Scholar

214. Wegner A, Elsenbruch S, Maluck J, Grigoleit JS, Engler H, Jager M, et al. Inflammation-induced hyperalgesia: effects of timing, dosage, and negative affect on somatic pain sensitivity in human experimental endotoxemia. Brain Behav Immun (2014) 41:46–54. doi:10.1016/j.bbi.2014.05.001

CrossRef Full Text | Google Scholar

215. Tracey I, Mantyh PW. The cerebral signature for pain perception and its modulation. Neuron (2007) 55:377–91. doi:10.1016/j.neuron.2007.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

216. Wegner A, Elsenbruch S, Rebernik L, Roderigo T, Engelbrecht E, Jager M, et al. Inflammation-induced pain sensitization in men and women: does sex matter in experimental endotoxemia? Pain (2015) 156(10):1954–64. doi:10.1097/j.pain.0000000000000256

CrossRef Full Text | Google Scholar

217. Sorge RE, Mapplebeck JC, 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):1081–3. doi:10.1038/nn.4053

PubMed Abstract | CrossRef Full Text | Google Scholar

218. Engler H, Benson S, Wegner A, Spreitzer I, Schedlowski M, Elsenbruch S. Men and women differ in inflammatory and neuroendocrine responses to endotoxin but not in the severity of sickness symptoms. Brain Behav Immun (2016) 52:18–26. doi:10.1016/j.bbi.2015.08.013

CrossRef Full Text | Google Scholar

219. Coyle SM, Calvano SE, Lowry SF. Gender influences in vivo human responses to endotoxin. Shock (2006) 26:538–43. doi:10.1097/01.shk.0000232589.39001.4d

PubMed Abstract | CrossRef Full Text | Google Scholar

220. Craig AD. Interoception: the sense of the physiological condition of the body. Curr Opin Neurobiol (2003) 13:500–5. doi:10.1016/S0959-4388(03)00090-4

CrossRef Full Text | Google Scholar

221. Benson S, Rebernik L, Wegner A, Kleine-Borgmann J, Engler H, Schlamann M, et al. Neural circuitry mediating inflammation-induced central pain amplification in human experimental endotoxemia. Brain Behav Immun (2015) 48:222–31. doi:10.1016/j.bbi.2015.03.017

PubMed Abstract | CrossRef Full Text | Google Scholar

222. Karshikoff B, Jensen KB, Kosek E, Kalpouzos G, Soop A, Ingvar M, et al. Why sickness hurts: a central mechanism for pain induced by peripheral inflammation. Brain Behav Immun (2016) 57:38–46. doi:10.1016/j.bbi.2016.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

223. Hannestad J, Subramanyam K, Dellagioia N, Planeta-Wilson B, Weinzimmer D, Pittman B, et al. Glucose metabolism in the insula and cingulate is affected by systemic inflammation in humans. J Nucl Med (2012) 53:601–7. doi:10.2967/jnumed.111.097014

PubMed Abstract | CrossRef Full Text | Google Scholar

224. Kullmann JS, Grigoleit JS, Lichte P, Kobbe P, Rosenberger C, Banner C, et al. Neural response to emotional stimuli during experimental human endotoxemia. Hum Brain Mapp (2013) 34:2217–27. doi:10.1002/hbm.22063

PubMed Abstract | CrossRef Full Text | Google Scholar

225. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Dolan RJ, et al. Neural origins of human sickness in interoceptive responses to inflammation. Biol Psychiatry (2009) 66:415–22. doi:10.1016/j.biopsych.2009.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

226. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry (2009) 66:407–14. doi:10.1016/j.biopsych.2009.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

227. Capuron L, Pagnoni G, Demetrashvili M, Woolwine BJ, Nemeroff CB, Berns GS, et al. Anterior cingulate activation and error processing during interferon-alpha treatment. Biol Psychiatry (2005) 58:190–6. doi:10.1016/j.biopsych.2005.03.033

PubMed Abstract | CrossRef Full Text | Google Scholar

228. Brydon L, Harrison NA, Walker C, Steptoe A, Critchley HD. Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry (2008) 63:1022–9. doi:10.1016/j.biopsych.2007.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

229. Inagaki TK, Muscatell KA, Irwin MR, Cole SW, Eisenberger NI. Inflammation selectively enhances amygdala activity to socially threatening images. Neuroimage (2012) 59:3222–6. doi:10.1016/j.neuroimage.2011.10.090

PubMed Abstract | CrossRef Full Text | Google Scholar

230. Dantzer R, Heijnen CJ, Kavelaars A, Laye S, Capuron L. The neuroimmune basis of fatigue. Trends Neurosci (2014) 37:39–46. doi:10.1016/j.tins.2013.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

231. Hannestad J, DellaGioia N, Ortiz N, Pittman B, Bhagwagar Z. Citalopram reduces endotoxin-induced fatigue. Brain Behav Immun (2011) 25:256–9. doi:10.1016/j.bbi.2010.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

232. Sandiego CM, Gallezot JD, Pittman B, Nabulsi N, Lim K, Lin SF, et al. Imaging robust microglial activation after lipopolysaccharide administration in humans with PET. Proc Natl Acad Sci U S A (2015). 112(40):12468–73. doi:10.1073/pnas.1511003112

CrossRef Full Text | Google Scholar

233. Hannestad J, Gallezot JD, Schafbauer T, Lim K, Kloczynski T, Morris ED, et al. Endotoxin-induced systemic inflammation activates microglia: [(1)(1)C]PBR28 positron emission tomography in nonhuman primates. Neuroimage (2012) 63:232–9. doi:10.1016/j.neuroimage.2012.06.055

CrossRef Full Text | Google Scholar

234. Brunson KL, Avishai-Eliner S, Hatalski CG, Baram TZ. Neurobiology of the stress response early in life: evolution of a concept and the role of corticotropin releasing hormone. Mol Psychiatry (2001) 6:647–56. doi:10.1038/sj.mp.4000942

PubMed Abstract | CrossRef Full Text | Google Scholar

235. Jankord R, Herman JP. Limbic regulation of hypothalamo-pituitary-adrenocortical function during acute and chronic stress. Ann N Y Acad Sci (2008) 1148:64–73. doi:10.1196/annals.1410.012

CrossRef Full Text | Google Scholar

236. Ulrich-Lai YM, Herman JP. Neural regulation of endocrine and autonomic stress responses. Nat Rev Neurosci (2009) 10:397–409. doi:10.1038/nrn2647

PubMed Abstract | CrossRef Full Text | Google Scholar

237. Bains JS, Wamsteeker Cusulin JI, Inoue W. Stress-related synaptic plasticity in the hypothalamus. Nat Rev Neurosci (2015) 16:377–88. doi:10.1038/nrn3881

PubMed Abstract | CrossRef Full Text | Google Scholar

238. Ji G, Fu Y, Adwanikar H, Neugebauer V. Non-pain-related CRF1 activation in the amygdala facilitates synaptic transmission and pain responses. Mol Pain (2013) 9:2. doi:10.1186/1744-8069-9-2

PubMed Abstract | CrossRef Full Text | Google Scholar

239. Su J, Tanaka Y, Muratsubaki T, Kano M, Kanazawa M, Fukudo S. Injection of corticotropin-releasing hormone into the amygdala aggravates visceral nociception and induces noradrenaline release in rats. Neurogastroenterol Motil (2015) 27:30–9. doi:10.1111/nmo.12462

PubMed Abstract | CrossRef Full Text | Google Scholar

240. Madden JT, Akil H, Patrick RL, Barchas JD. Stress-induced parallel changes in central opioid levels and pain responsiveness in the rat. Nature (1977) 265:358–60. doi:10.1038/265358a0

CrossRef Full Text | Google Scholar

241. Willer JC, Dehen H, Cambier J. Stress-induced analgesia in humans: endogenous opioids and naloxone-reversible depression of pain reflexes. Science (1981) 212:689–91. doi:10.1126/science.6261330

PubMed Abstract | CrossRef Full Text | Google Scholar

242. Paananen M, O’Sullivan P, Straker L, Beales D, Coenen P, Karppinen J, et al. A low cortisol response to stress is associated with musculoskeletal pain combined with increased pain sensitivity in young adults: a longitudinal cohort study. Arthritis Res Ther (2015) 17:355. doi:10.1186/s13075-015-0875-z

PubMed Abstract | CrossRef Full Text | Google Scholar

243. Sudhaus S, Fricke B, Stachon A, Schneider S, Klein H, von During M, et al. Salivary cortisol and psychological mechanisms in patients with acute versus chronic low back pain. Psychoneuroendocrinology (2009) 34:513–22. doi:10.1016/j.psyneuen.2008.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

244. Vachon-Presseau E, Roy M, Martel MO, Caron E, Marin MF, Chen J, et al. The stress model of chronic pain: evidence from basal cortisol and hippocampal structure and function in humans. Brain (2013) 136:815–27. doi:10.1093/brain/aws371

CrossRef Full Text | Google Scholar

245. Hannibal KE, Bishop MD. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys Ther (2014) 94:1816–25. doi:10.2522/ptj.20130597

PubMed Abstract | CrossRef Full Text | Google Scholar

246. Zell V, Juif PE, Hanesch U, Poisbeau P, Anton F, Darbon P. Corticosterone analgesia is mediated by the spinal production of neuroactive metabolites that enhance GABAergic inhibitory transmission on dorsal horn rat neurons. Eur J Neurosci (2015) 41:390–7. doi:10.1111/ejn.12796

PubMed Abstract | CrossRef Full Text | Google Scholar

247. Asnis GM, De La Garza R II. Interferon-induced depression in chronic hepatitis C: a review of its prevalence, risk factors, biology, and treatment approaches. J Clin Gastroenterol (2006) 40:322–35. doi:10.1097/01.mcg.0000210099.36500.fe

PubMed Abstract | CrossRef Full Text | Google Scholar

248. Raison CL, Miller AH. Role of inflammation in depression: implications for phenomenology, pathophysiology and treatment. Mod Trends Pharmacopsychiatri (2013) 28:33–48. doi:10.1159/000343966

PubMed Abstract | CrossRef Full Text | Google Scholar

249. Danese A, Pariante CM, Caspi A, Taylor A, Poulton R. Childhood maltreatment predicts adult inflammation in a life-course study. Proc Natl Acad Sci U S A (2007) 104:1319–24. doi:10.1073/pnas.0610362104

PubMed Abstract | CrossRef Full Text | Google Scholar

250. Dantzer R, O’Connor JC, Lawson MA, Kelley KW. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology (2011) 36:426–36. doi:10.1016/j.psyneuen.2010.09.012

CrossRef Full Text | Google Scholar

251. Grimby-Ekman A, Gerdle B, Bjork J, Larsson B. Comorbidities, intensity, frequency and duration of pain, daily functioning and health care seeking in local, regional, and widespread pain – a descriptive population-based survey (SwePain). BMC Musculoskelet Disord (2015) 16:165. doi:10.1186/s12891-015-0631-1

CrossRef Full Text | Google Scholar

252. Fernandez M, Ordonana JR, Hartvigsen J, Ferreira ML, Refshauge KM, Sanchez-Romera JF, et al. Is chronic low back pain associated with the prevalence of coronary heart disease when genetic susceptibility is considered? A co-twin control study of Spanish twins. PLoS One (2016) 11:e0155194. doi:10.1371/journal.pone.0155194

PubMed Abstract | CrossRef Full Text | Google Scholar

253. Lasselin J, Capuron L. Chronic low-grade inflammation in metabolic disorders: relevance for behavioral symptoms. Neuroimmunomodulation (2014) 21:95–101. doi:10.1159/000356535

PubMed Abstract | CrossRef Full Text | Google Scholar

254. Burke NN, Finn DP, McGuire BE, Roche M. Psychological stress in early life as a predisposing factor for the development of chronic pain: clinical and preclinical evidence and neurobiological mechanisms. J Neurosci Res (2016). doi:10.1002/jnr.23802

CrossRef Full Text | Google Scholar

255. DeVon HA, Piano MR, Rosenfeld AG, Hoppensteadt DA. The association of pain with protein inflammatory biomarkers: a review of the literature. Nurs Res (2014) 63:51–62. doi:10.1097/NNR.0000000000000013

PubMed Abstract | CrossRef Full Text | Google Scholar

256. Puntener U, Booth SG, Perry VH, Teeling JL. Long-term impact of systemic bacterial infection on the cerebral vasculature and microglia. J Neuroinflammation (2012) 9:146. doi:10.1186/1742-2094-9-146

PubMed Abstract | CrossRef Full Text | Google Scholar

257. Drake C, Boutin H, Jones MS, Denes A, McColl BW, Selvarajah JR, et al. Brain inflammation is induced by co-morbidities and risk factors for stroke. Brain Behav Immun (2011) 25:1113–22. doi:10.1016/j.bbi.2011.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

258. Ohmoto Y, Wood MJ, Charlton HM, Kajiwara K, Perry VH, Wood KJ. Variation in the immune response to adenoviral vectors in the brain: influence of mouse strain, environmental conditions and priming. Gene Ther (1999) 6:471–81. doi:10.1038/sj.gt.3300851

CrossRef Full Text | Google Scholar

259. Cunningham C, Campion S, Lunnon K, Murray CL, Woods JF, Deacon RM, et al. Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychiatry (2009) 65:304–12. doi:10.1016/j.biopsych.2008.07.024

PubMed Abstract | CrossRef Full Text | Google Scholar

260. Cunningham C, Wilcockson DC, Campion S, Lunnon K, Perry VH. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J Neurosci (2005) 25:9275–84. doi:10.1523/JNEUROSCI.2614-05.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

261. Bugada D, Lavand’homme P, Ambrosoli AL, Cappelleri G, Saccani Jotti GM, Meschi T, et al. Effect of preoperative inflammatory status and comorbidities on pain resolution and persistent postsurgical pain after inguinal hernia repair. Mediators Inflamm (2016) 2016:5830347. doi:10.1155/2016/5830347

PubMed Abstract | CrossRef Full Text | Google Scholar

262. Li Y, Oosting M, Deelen P, Ricano-Ponce I, Smeekens S, Jaeger M, et al. Inter-individual variability and genetic influences on cytokine responses to bacteria and fungi. Nat Med (2016) 22:952–60. doi:10.1038/nm1016-1192b

CrossRef Full Text | Google Scholar

263. Qu K, Zaba LC, Giresi PG, Li R, Longmire M, Kim YH, et al. Individuality and variation of personal regulomes in primary human T cells. Cell Syst (2015) 1:51–61. doi:10.1016/j.cels.2015.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

264. Brodin P, Jojic V, Gao T, Bhattacharya S, Angel CJ, Furman D, et al. Variation in the human immune system is largely driven by non-heritable influences. Cell (2015) 160:37–47. doi:10.1016/j.cell.2014.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

265. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol (2011) 11:519–31. doi:10.1038/nri3024

CrossRef Full Text | Google Scholar

266. Vos R, van den Akker M, Boesten J, Robertson C, Metsemakers J. Trajectories of multimorbidity: exploring patterns of multimorbidity in patients with more than ten chronic health problems in life course. BMC Fam Pract (2015) 16:2. doi:10.1186/s12875-014-0213-6

PubMed Abstract | CrossRef Full Text | Google Scholar

267. Perry VH. Contribution of systemic inflammation to chronic neurodegeneration. Acta Neuropathol (2010) 120:277–86. doi:10.1007/s00401-010-0722-x

CrossRef Full Text | Google Scholar

268. Chen MH, Li CT, Tsai CF, Lin WC, Chang WH, Chen TJ, et al. Risk of dementia among patients with asthma: a nationwide longitudinal study. J Am Med Dir Assoc (2014) 15:763–7. doi:10.1016/j.jamda.2014.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

269. Rusanen M, Ngandu T, Laatikainen T, Tuomilehto J, Soininen H, Kivipelto M. Chronic obstructive pulmonary disease and asthma and the risk of mild cognitive impairment and dementia: a population based CAIDE study. Curr Alzheimer Res (2013) 10:549–55. doi:10.2174/1567205011310050011

PubMed Abstract | CrossRef Full Text | Google Scholar

270. Davies MJ, Norman RJ. Programming and reproductive functioning. Trends Endocrinol Metab (2002) 13:386–92. doi:10.1016/S1043-2760(02)00691-4

PubMed Abstract | CrossRef Full Text | Google Scholar

271. Welberg LA, Seckl JR. Prenatal stress, glucocorticoids and the programming of the brain. J Neuroendocrinol (2001) 13:113–28. doi:10.1111/j.1365-2826.2001.00601.x

PubMed Abstract | CrossRef Full Text | Google Scholar

272. Del Giudice M. Fetal programming by maternal stress: insights from a conflict perspective. Psychoneuroendocrinology (2012) 37:1614–29. doi:10.1016/j.psyneuen.2012.05.014

PubMed Abstract | CrossRef Full Text | Google Scholar

273. Gluckman PD, Hanson MA, Spencer HG. Predictive adaptive responses and human evolution. Trends Ecol Evol (2005) 20:527–33. doi:10.1016/j.tree.2005.08.001

CrossRef Full Text | Google Scholar

274. Minde JK. Norrbottnian congenital insensitivity to pain. Acta Orthop Suppl (2006) 77:2–32. doi:10.1080/17453690610046495a

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: psychoneuroimmunology, lipopolysaccharide, inflammation, pain, neuroimmunology, neuroendocrinology, hypothalamo–pituitary–adrenal axis, stress

Citation: Zouikr I and Karshikoff B (2017) Lifetime Modulation of the Pain System via Neuroimmune and Neuroendocrine Interactions. Front. Immunol. 8:276. doi: 10.3389/fimmu.2017.00276

Received: 07 October 2016; Accepted: 24 February 2017;
Published: 13 March 2017

Edited by:

Hans-Peter Hartung, University of Düsseldorf, Germany

Reviewed by:

Samar S. Ayache, Paris Est University Creteil, France
Anna Fogdell-Hahn, Karolinska Institutet, Sweden

Copyright: © 2017 Zouikr and Karshikoff. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ihssane Zouikr, ihssane.zouikr@uon.edu.au

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.