Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 10.
Published in final edited form as: Nat Rev Immunol. 2015 May 15;15(6):335–349. doi: 10.1038/nri3843

Fever and the thermal regulation of immunity: the immune system feels the heat

Sharon S Evans 1, Elizabeth A Repasky 1, Daniel T Fisher 1
PMCID: PMC4786079  NIHMSID: NIHMS755275  PMID: 25976513

Abstract

Fever is a cardinal response to infection that has been conserved in warm and cold-blooded vertebrates for over 600 million years of evolution. The fever response is executed by integrated physiological and neuronal circuitry and confers a survival benefit during infection. Here, we review our current understanding of how the inflammatory cues delivered by the thermal element of fever stimulate innate and adaptive immune responses. We further highlight the unexpected multiplicity of roles of the pyrogenic cytokine interleukin-6 (IL-6), both during fever induction as well as during the mobilization of lymphocytes to the lymphoid organs that are the staging ground for immune defence. Finally, we discuss the emerging evidence that suggests the adrenergic signalling pathways associated with thermogenesis shape immune cell function.


The fever response is a hallmark of infection and inflammatory disease and has been shaped through hundreds of millions of years of natural selection. Febrile temperatures are so closely linked to the inflammatory response that heat (calor) is one of the four cardinal signs of inflammation, along with pain (dolor), redness (rubor), and swelling (tumour), as described by Celsus in ~30 BC.1 The induction of fever in endothermic (warm-blooded) animals occurs at a high metabolic cost such that a 1°C rise in body temperature requires a 10–12.5% increase in metabolic rate.2 There is mounting evidence that the increase of 1 to 4°C in core body temperature that occurs during fever is associated with improved survival and resolution of many infections. For example, the use of antipyretic drugs to diminish fever correlates with a 5% increase in mortality in human populations infected with influenza virus and negatively affects patient outcomes in the intensive care unit.35 Preclinical studies in rabbits infected with rinderpest virus also found an increase in mortality when fever was inhibited with the antipyretic drug acetylsalicylic acid — 70% of acetylsalicylic acid-treated animals died as a result of infection as compared with only 16% of animals with a normal febrile response.6 However, fever is not universally beneficial, particularly in cases of extreme inflammation where lowering, rather than raising body temperature has evolved as a protective mechanism.710 Thus, uncontrolled fever is associated with worse outcomes in patients with sepsis or neurological injuries, whereas treatments that induce hypothermia can have a clinical benefit.11,12 A challenge in ascertaining the precise value of fever in endotherms is that the antipyretics used to inhibit fever target multiple aspects of the inflammatory response besides temperature regulation.11

Ectothermic (cold-blooded) vertebrates, which last shared a common ancestor with mammals over 600 million years ago, provide an ‘experiment in nature’ in which to examine the direct impact of febrile temperatures on survival. Ectotherms as diverse as reptiles, fish, and insects raise their core temperature during infection through behavioural regulation, which leads to their seeking warmer environments (despite the risk of predation) or, in the case of bees, raising the local temperature of the hive through increased physical activity.2,1319 Landmark studies published 40 years ago by Kluger’s laboratory showed that survival of the desert iguana Dipsosaurus dorsalis is reduced by 75% if prevented from behaviourally raising its core temperature by approximately 2°C after infection with the Gram-negative bacterium Aeromonas hydrophila.2,13,14 The heat-seeking behaviour of the desert iguana, blue-finned tuna and leech is negated by antipyretic drugs, indicating that common biochemical pathways drive fevers in ectothermic and endothermic animals.14,16,20 Surprisingly, the correlation between infection and increased temperature extends even to plants, which arose 1.5 billion years ago. For example, the temperature of the leaves from the bean plant Phaseolus vulgaris increases by around 2°C following infection with the fungus Collectotrichum lindemuthianum.21 Thermoregulation in plants occurs through mitochondrial respiration22, although it is not known whether these fever-like responses have a direct impact on clearance of infection.

The fact that fever has been retained throughout vertebrate evolution strongly argues that febrile temperatures confer a survival advantage. A long-standing mystery relates to the protective mechanisms by which fever wards off attacks by invading pathogens. One mechanism involves direct effects of febrile temperatures on the infectious potential of pathogens.23 For example, temperatures in the febrile range (40–41°C) cause a greater than 200-fold reduction in the replication rate of poliovirus in mammalian cells and increase the susceptibility of Gram-negative bacteria to serum-induced lysis.24,25 In this Review, we discuss the evidence suggesting that febrile temperatures boost the effectiveness of the immune response during infections by stimulating both the innate and adaptive arms of the immune system. We will highlight the role of the pyrogenic cytokine interleukin-6 (IL-6) in two key phases of the febrile response: firstly in driving the rise in core temperature, and secondly as a downstream effector cytokine orchestrating lymphocyte trafficking to lymphoid organs. We also describe febrile temperature as a ‘rheostat’, dialling down systemic inflammation during the return to homeostasis. Finally, we highlight new data demonstrating the overlapping signalling pathways that are involved in thermogenesis and in the regulation of the immune response. We only briefly discuss the neuronal circuitry that drives fever and the evolutionarily conserved heat shock protein (HSP) response (BOX 1), but refer the reader to recent comprehensive reviews for additional information on these topics as well as on the contributions of hypothermia to limiting inflammation.2630

BOX 1. Thermal regulation of heat shock proteins.

Heat shock proteins (HSP) are cytoprotective proteins that are constitutively expressed and also rapidly induced under proteotoxic stress conditions such as heat, hypoxia, oxidative stress, toxin exposure, nutrient deprivation, and infection.2629,200202 Although HSPs were originally discovered in the context of heat shock (42–45°C), they are also inducible by febrile temperatures in mammalian cells (38–41°C).26,122,189191 Stress-induced transcription of HSPs is driven by post-translational modifications (sumoylation and phosphorylation) of heat shock factor protein 1 (HSF1) which release it from a complex with HSP70 and HSP90.29,200,201 This results in the formation of HSF1 homotrimers that translocate to the nucleus and activate transcription of genes including HSPs that contain ‘heat shock element’ sequences.29,200,201 The major function of HSPs is to maintain appropriate folding of their client proteins, thereby protecting them from proteolysis. HSPs have key roles in regulating multiple signalling pathways under constitutive and stress conditions. For example, there are more than 200 established client proteins of HSP90, including members of the MAPK, JAK–STAT and CDK1 signalling pathways.29,200,202204 Cancer cells under chronic proteotoxic stress conditions often become ‘addicted’ to HSPs and high intratumoral expression of HSP70 or HSP90 is a poor prognostic indicator in cancer patients, suggesting HSP inhibitors as a treatment option in cancer.200,203,204 There are also non-canonical HSPs that do not have traditional chaperoning/protein folding activity but whose expression is nonetheless tightly regulated by HSF1. One example is the chemokine CXCL8 (also known as IL-8) that mediates recruitment of neutrophils upon exposure to fever-range hyperthermia in LPS instillation models of acute lung inflammation.84,85 Active areas of investigation in the HSP field are considering the physiological impact of the multiple post-translational modifications of HSFs and HSPs (for example, phosphorylation, acetylation, S-nitrosylation, ubiquitination, and sumoylation) as well as the interplay between these molecules and positive and negative immune regulation.29,200,205

Induction of fever

The IL-6–COX2–PGE2 axis drives fever

The induction and maintenance of fever during infection involves the tightly coordinated interplay between the innate immune system and neuronal circuitry within the central and peripheral nervous systems. Immune sensing of infection begins with the binding of pathogen-associated molecular patterns (for example, lipopolysaccharide ((LPS)), viral RNA, or fungal sugars) to pathogen recognition receptors (PRRs), such as Toll-like receptors (TLs), which are expressed by innate immune cell populations, including macrophages, neutrophils and dendritic cells (DCs) (FIG. 1). Much of our current understanding of the molecular mechanisms underlying fever stems from studies in which rodents were injected with LPS, a component of Gram-negative bacterial cell walls, to model immune-induced thermoregulation. In this model, prostaglandin E2 (PGE2) produced by brain vascular endothelial cells is considered a major pyrogenic mediator of fever.3133 This lipid effector molecule integrates input signals from pyrogenic cytokines produced in response to pathogenic stimuli, with output signals involving neurotransmitters that raise core body temperature (FIG. 1). PGE2 is also synthesized in the periphery early in this response – that is, prior to the detection of circulating cytokines. It is produced by hematopoietic cells following LPS-mediated activation of TLR4 and travels through the blood/brain barrier to initiate fever.26,30,3438 LPS-induced fever occurs via autonomic mechanisms driven by PGE2 binding to EP3 prostaglandin receptors expressed by thermoregulatory neurons in the median preoptic nucleus within the hypothalamus.8,3941 Endotherms elevate body temperature through the release of norepinephrine, which increases thermogenesis in brown adipose tissue and induces vasoconstriction in the extremities to reduce passive heat loss.26,27 In addition, signalling through the neurotransmitter acetylcholine stimulates the musculature to convert stored chemical energy into thermal energy and increases overall metabolic rates.2,26,42,43 Endotherms, like ectotherms, also engage in heat-seeking behavioral thermoregulation which does not require median preoptic neurons although the pathways involved are largely unknown.810

Figure 1. The induction of fever during infection.

Figure 1

The recognition of damage-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS), by Toll-like receptors (TLRs) and other pattern recognition receptors drives the activation of dendritic cells (DCs) and macrophages. These innate immune cells release prostaglandin E2 (PGE2) as well as pyrogenic cytokines (namely, interleukin-1 (IL-1) IL-6, and tumour necrosis factor (TNF)) that act systemically to induce fever. IL-6 operates downstream of IL-1 in the median preoptic nucleus region within the hypothalamus to induce the synthesis of cyclooxygenase 2 (COX2), the enzyme responsible for production of additional PGE2.64,65 PGE2 is considered the major pyrogenic mediator of fever.3133 Receptor activator of NF-κB (RANK) expressed by astrocytes also acts via the COX2–PGE2 pathway to induce fever.47 However, it is not known whether this pathway parallels the IL-6 response or if the IL-6 and RANKL pathways converge, potentially via IL-6 regulation of RANKL expression in vascular endothelial cells in the hypothalamus. Neurons expressing PGE2 receptor 3 (EP3) trigger the sympathetic nervous system to trigger norepinephrine release, which elevates body temperature by increasing thermogenesis in brown adipose tissue as well as by inducing vasoconstriction to prevent passive heat loss.2,26,27,42,43 Additionally, acetylcholine contributes to fever by stimulating muscle myocytes to induce shivering.

LPS-induced TLR4 signalling stimulates the synthesis of pyrogenic cytokines (namely IL-1, tumour necrosis factor ((TNF)) and IL-6) at the site of infection as well as within the brain, and it is becoming clear that IL-6 is as an important mediator of fever induction.26,4447 Notably, multiple cell types within the brain (for example, astrocytes, microglial cells and neurons) have the capacity to synthesize IL-6 in response to local inflammatory stimuli4853. Although the direct administration of TNF, IL-1, or IL-6 into the brain leads to a febrile response, several lines of evidence point to a requisite role for IL-6 in sustaining fever. In this regard, LPS-induced fever does not occur in the presence of IL-6-specific neutralizing antibody or in IL-6-deficient mice, even though TNF and IL-1 upregulation is normal in these settings.5458 Moreover, direct intracerebroventricular injection of IL-6, but not IL-1, restores febrile responses in IL-6-deficient mice.55 Febrile temperatures have further been implicated in a positive feedback loop during the early stages of infection. Specifically, passive elevation of the core body temperature of mice to the febrile range using whole body hyperthermia substantially augments circulating levels of TNF, IL-1, and IL-6 during LPS-induced inflammation.26,5961 The pyrogenic role of IL-6 has recently been corroborated in patients with paediatric leukaemia, where treatment with the IL-6 receptor antagonist tocilizumab was found to reverse the high fevers that develop during T cell based-immunotherapy (specifically, following the administration of chimeric antigen receptor-expressing T cells or a CD19/CD3-bispecific antibody).62,63

Systemic or locally produced cytokines act in the brain to augment the synthesis of cyclooxygenase 2 (COX2), the enzyme responsible for oxidizing arachidonic acid to produce PGE2 (FIG. 1). For instance, IL-1 receptors that mediate COX2 induction have been identified on brain endothelial cells within the preoptic region of the hypothalamus.64,65 Although the specific cell types that upregulate COX2 in response to IL-6 remain to be identified, blood vessels in the brain reportedly express the IL-6 receptor-α subunit,53 which together with the ubiquitously expressed gp130 subunit forms the functional IL-6 receptor. Several studies have shown that cerebral COX2, PGE2, and fever are not induced during LPS-driven inflammation in IL-6-deficient mice or in the presence of IL-6-specific neutralizing antibody.6668 Alternatively, IL-6 cannot initiate a febrile response in the absence of COX2 or PGE2, and intracerebroventricular delivery of PGE2 bypasses the requirement of IL-6 for fever induction in IL-6-deficient mice.69,70 Collectively, these observations establish that COX2 and PGE2 are crucial mediators that can operate downstream of IL-6 in the LPS-induced febrile response.

RANKL and fever induction

An open question is whether IL-6 is the direct regulator of COX2 and PGE2 induction during the febrile response or whether other intervening cytokines are involved. The latter possibility is suggested by an elegant study by Hanada et al. showing that, similarly to IL-6, the cytokine known as receptor-activator of NF-kB ligand (RANKL) converges on the COX2–EP3–PGE2 pathway leading to fever induction in the LPS-induced model of inflammation47 (FIG. 1). RANKL is best known as a regulator of bone remodelling and lymph node organogenesis.71 However, mRNA encoding RANKL is also produced in the lateral septal nucleus region of the brain that interconnects with the hypothalamus and the RANKL receptor, RANK, is found on astrocytes in the preoptic region of the hypothalamus.47 Further support for a role of this cytokine in thermoregulation is provided by findings that children with RANK mutations exhibit impaired fever responses during pneumonia.47 Although the potential interplay between IL-6 and RANKL–RANK during fever has not been explored, it is tempting to speculate that RANKL is a downstream mediator of IL-6-induced pyrogenesis based on evidence that IL-6 directly stimulates RANKL synthesis by synovial fibroblasts in mouse models of rheumatoid arthritis.72

Immune stimulation by thermal stress

One benefit widely attributed to fever is the enhancement of immune-protective mechanisms during infection. Defence against pathogens involves tight spatial and temporal regulation of the immune system, and the same pyrogenic cytokines that are produced during the induction of fever also operate locally to orchestrate immunity within infected tissues.73 Innate immune cells are the ‘first responders’, arriving within hours to directly destroy pathogens via phagocytic or cytotoxic activities. These activities limit infection until a peak adaptive immune response is generated, normally around one week later. Macrophages and DCs bridge the gap between innate and adaptive immunity by taking up pathogens in peripheral tissues and then relocating to draining lymph nodes where they drive expansion of pathogen-specific effector T cells.74,75 Crucial for this process is the co-localization of DCs and T cells near high endothelial venules (HEVs) that are the major portals for entry of blood-borne lymphocytes.7476

Given the complexity of these immune mechanisms, it is remarkable that fever-range temperatures stimulate almost every step involved in this process, promoting both innate and adaptive immunity. The potential impact of the thermal element of fever has primarily been explored using hyperthermic temperatures within the febrile range for mammals (that is, ranging from 38–41°C; ΔT~1–4°C above baseline) in the various in vitro and in vivo studies described below. Experimental hyperthermia is a powerful approach to study the impact of febrile-range temperatures on immunity, which is otherwise difficult to discriminate during natural fever because of the attendant inflammatory programme (comprised of lipid and cytokine mediators) that regulate both fever and immunity. However, an important caveat from a physiological perspective, is that the heat conservation associated with natural fever differs fundamentally from cooling mechanisms enacted by thermoregulation following exogenous heat application.

Impact of febrile temperatures on innate immunity

Previous research using animal models of hyperthermia treatment alone, or with LPS challenge or bacterial infection, strongly supports the idea that fever-range temperatures elevate the respiratory burst typically associated with activation and bacteriolytic activity of neutrophils77,78 (FIG. 2a). Thermal stress further increases neutrophil recruitment to local sites of infection and other distant tissues61,79 (FIG. 2a) including tumours77 Neutrophil localization in peripheral tissues is due, at least in part, to heat-induced increases in circulating neutrophils which are dependent upon granulocyte colony-stimulating factor (G-CSF).80,81 G-CSF is also central in a model of radiation-induced neutropenia where fever-range whole body hyperthermia substantially increases the rate of neutrophil recovery in the blood, and augments the number hematopoietic stem cells and neutrophil progenitors within the bone marrow82 (FIG. 2a). This effect is dependent upon enhanced production of IL-17, IL-1β and IL-1α preferentially in intestinal tissue. Importantly, the precise outcome of the thermal effect depends on the heating protocol used and the geography of cell recruitment (FIG. 2a). Indeed, temperatures above the normal febrile range impair neutrophil accumulation and function.83 Moreover, Hasday and colleagues found that fever, or exposure to fever-range hyperthermia, in an LPS model increases neutrophil localization to the lung, which can have negative consequences due to inflammation-induced local tissue damage.61,84 Heat-induced neutrophil recruitment in the lung depends on the non-canonical chemotactic HSP, CXCL8 (also known as IL-8), which is expressed under the control of the heat-inducible transcription factor heat shock factor 1 (HSF1) (BOX 1).61,84,85 Neutrophil recruitment in the lung also involves a decrease in endothelial barrier integrity through a mechanism depending on p38 MAPK and ERK1–ERK2 signalling.84

Figure 2. Response of innate immune cells to thermal stress.

Figure 2

(a) Fever-range temperatures drive several crucial aspects of innate immunity. Fever-range hyperthermia stimulates the release of neutrophils from the bone marrow in a granulocyte–colony-stimulating factor (G-CSF)-driven manner.8082 Febrile-range temperatures also promote neutrophil recruitment to the lungs and other local sites of infection in a CXC-chemokine ligand 8 (CXCL8)-dependent fashion that additionally involves decreased barrier function of vessels.61,84,85 Upon arriving in the site of infection, thermal stress further elevates the respiratory burst which increases the bacteriolytic activity of neutrophils.77,78 (b) Thermal treatment improves natural killer (NK) cell cytolytic activity through induction of MHC class I polypeptide-related sequence A (MICA) expression on target cells (for example, tumour cells) as well as by inducing the clustering of the MICA counter-receptor NKG2D on the surface of NK cells.90 (c) Temperatures in the febrile range increase the ability of antigen-presenting cells to support the formation of the adaptive immune response. Heat improves the phagocytic potential of macrophages and dendritic cells (DCs) and increases their responsiveness to invading pathogens by upregulating their expression of both Toll-like receptor 2 (TLR2) and TLR4.119,120 Thermal treatment also induces the release of immunomodulatory molecules such as cytokines (for example, TNF), nitric oxide (NO) and heat shock protein 70 (HSP70). Additionally, heat increases expression of MHC class I and II molecules as well as co-stimulatory molecules (CD80 and CD86) by mature DCs and augments their CC-chemokine receptor 7 (CCR7)-dependent migration via the afferent lymphatics that serve as a conduit to draining lymph nodes.117,121124 DCs exposed to febrile temperatures are also more efficient at cross-presenting antigens and inducing T helper 1 (Th1) cell polarization.121

The impact of heat on natural killer (NK) cells has been most extensively studied in the context of tumour immunity. It has been shown that NK cell cytotoxic activity and recruitment to tumour sites is increased by fever-range hyperthermia in vivo8689 (FIG. 2b). This enhanced cytotoxicity depends upon heat-induced upregulation of the NKG2D ligand MICA (MHC class I polypeptide-related sequence A) on tumour cells as well as on the clustering of NKG2D receptors on the surface of NK cells.90 Elevated temperatures also decrease MHC class I expression by tumour cells while simultaneously increasing HSP70 production, and both of these responses are linked to enhanced cytotoxic potential in NK cells.91 The upregulation of HSPs in tumour cells in response to thermal stress is also likely to be involved in the enhanced cross-priming of antigen-specific cytotoxic T lymphocytes that was observed when DCs were loaded with lysate from heated melanoma cells.92

Macrophages have served as a major model for the study of febrile-range hyperthermia. Early studies demonstrated that whole-body heating (to ~39.5°C) improves bacterial clearance and also increases serum concentrations of TNF, IL-1, and IL-6 in mice challenged with LPS.59,60,93,94 The source of these cytokines was found to be the macrophages of the liver (that is, Kupffer cells) as well as macrophages in other organs. Later work by Lee et al. showed that hyperthermia induces the upregulation of HSP70 and this ‘reprogrammes’ macrophages to show sustained activation in response to LPS.95 The mechanism involves the phosphorylation of the IKK and IκB kinases, the nuclear translocation of NFκB and its binding to the Tnfpromoter.95,96 HSP70 is also required for enhancing the expression of nitric oxide and inducible nitric oxide synthase by peritoneal macrophages following exposure to fever-range temperatures together with LPS and IFNγ.97 Although HSPs are usually assumed to be intracellular, heat stress can induce HSP70 release from cells into the extracellular environment where it can act as a damage-associated molecular pattern (DAMP) to stimulate macrophages and DCs.98100 Extracellular HSP70 and other HSPs engage multiple surface receptors, including CD91, scavenger receptor A, CD40, TLR2 or TLR4, leading to the release of nitric oxide, TNF, IL-6, IL-1β and IL-12.100110 Of note, some investigators have paradoxically observed an anti-inflammatory role for HSPs.111113 It is suggested that these differences result from the precise location of HSPs within macrophages: extracellular HSPs provide danger signals to enhance inflammation whereas intracellular HSP could help to suppress inflammatory signalling.114 Taken together, the data regarding innate immune cells, body temperature and HSPs reveal fascinating, yet still poorly understood, layers of interdependency between the febrile response and the more ancient HSP response.

Fever enhances DC functions

Several studies demonstrate that elevated temperatures substantially enhance the phagocytic potential of DCs, in addition to augmenting interferon-α (IFNα) production in response to viral infection (FIG. 2c).115118 Heating of immature DCs also upregulates their expression of TLR2 and TLR4, suggesting a role for thermal signals in enhancing pathogen sensing by innate immune cells.119,120 Febrile temperatures further increase DC expression of MHC class I and class II molecules and co-stimulatory molecules, including CD80 and CD86, and can augment the secretion of the Th1 cell-polarizing cytokines IL-12 and TNF.102,117,119123 Additional reports point to a role for febrile-range temperatures in augmenting the migration of antigen-presenting cells (APCs), such as skin Langerhans cells, to draining lymph nodes124 (FIG. 2c). These data may help to explain the fact that febrile temperatures can accelerate the swelling phase of a contact hypersensitivity reaction when heat is delivered to mice shortly after the application of the elicitation dose of a skin sensitizer, fluorescein isothiocyanate (FITC).124 The underlying mechanism directing DC migration to draining lymph nodes likely involves increased responsiveness of CC-chemokine receptor 7 (CCR7) to its ligands, which has been described for heat-treated mature DCs in chemotaxis assays in vitro.121 CCR7 senses CCL21 chemokine gradients in vivo, thereby guiding DC entry into afferent lymphatics and their subsequent migration near HEVs within draining nodes.125128 Thus, febrile temperatures appear to regulate the CCR7–CCL21 axis in order to optimally position DCs in lymphoid organs at sites where they can present antigen to lymphocytes upon their arrival via HEVs.

Given these observations, it is not surprising that fever-range thermal stress enhances the ability of DCs to stimulate T cells as well as DC cross-presenting functions (FIG. 2c). In mixed lymphocyte reactions, applying thermal stress ex vivo to LPS-pulsed mature human monocyte-derived DCs led to enhanced proliferation of naïve CD4+ T cells and promoted their differentiation towards a Th1 cell phenotype.121 Similarly, DCs isolated from heated mice exhibit a superior ability to activate T cells.102 In studies where DCs from patients with medullary thyroid cancer were preheated prior to co-culture with T cells, the T cells showed enhanced cytotoxicity against tumour targets.119 This increased cytotoxicity of effector T cells correlated with heat-induced upregulation of both MHC class I and HSP70 expression in mature, but not immature, DCs. Together, these findings demonstrate that systemic fever-range temperatures can target different components of the innate immune system, including the HSP response, in order to enhance effector T cell responses.

Thermal mechanisms boost adaptive immunity

A crucial determinant for the generation of adaptive immunity is the high rate of lymphocyte trafficking through lymphoid organs. The entire pool of naive T cells in a mouse lymph node turn overs ~2–3 times per day as a result of T cell recirculation.75,129 This dynamic flux increases the probability that rare antigen-specific T cells (present at a frequency of only ~1 in 105–106)130,131 will receive activating signals from DCs. The entry of blood-borne T and B cells into lymph nodes and Peyer’s patches occurs preferentially at HEVs through a well-defined adhesion cascade that involves; one, L-selectin and/or α4β7 integrin initiated tethering and rolling; two, CCL21-dependent activation of CCR7 on adherent lymphocytes; three, LFA1–mediated firm arrest via binding to its endothelial counter-receptors intercellular adhesion molecule-1 (ICAM1) and ICAM2; and four, LFA1–ICAM1–2-directed transendothelial migration.7476,132,133 As described below, we have shown that fever-range thermal stress targets multiple steps in this cascade by invoking a wide array of lymphocyte and endothelial trafficking molecules134142 (FIG. 3a).

Figure 3. Fever-range thermal stress and the adaptive immune response.

Figure 3

(a) Fever-range thermal stress supports increased adaptive immunity by targeting two distinct aspects of T cell activation in lymph nodes. Heat enhances the rate of lymphocyte trafficking across high endothelial venules (HEVs) in peripheral lymph nodes through effects on each step of the adhesion cascade. Heat treatment of lymphocytes increases the frequency of L-selectin-dependent tethering and rolling interactions.134,135,137139 Febrile-range temperatures independently act on HEVs to enhance the transition of lymphocytes from transient rolling to stable arrest by increasing the intravascular density of CC-chemokine ligand 21 (CCL21) and intracellular adhesion molecule 1 (ICAM1).140142 ICAM1 also supports lymphocyte crawling to inter-endothelial cell junctions as well as transendothelial migration.131,145,146 Heat also acts directly on the T cells within lymphoid organs by pre-clustering components of the immunological synapse (TCRβ and CD8) into lipid rafts. This prolongs stable contacts with APCs and increases CD8+ T cell differentiation towards an effector phenotype characterized by enhanced L-selectin downregulation, cytotoxic function, and production of interferon-γ (IFNγ).151,152 (b) Epifluorescence whole-mount confocal microscopy imaging of HEVs that are actively supporting lymphocyte trafficking in a mouse lymph node. HEVs are stained in red with PE-conjugated MECA-79 antibody that recognizes peripheral lymph node addressin (PNAD) whereas lymphocytes are labelled in green using carboxyfluorescein succinimidyl ester (CFSE).

An early indication that fever could control lymphocyte trafficking emerged from studies showing transient decreases in circulating T cells in mice or patients with cancer following elevation of core body temperatures to ~39.5°C by febrile-range whole body hyperthermia.83,137,143 Reductionist studies found that direct heat treatment of T or B cells ex vivo for 6 hours resulted in an approximately 2-fold increase in their ability to bind to HEVs in vitro or to home to lymph nodes or Peyer’s patches in vivo.134139 Lymphocytes isolated from heated mice exhibit similar enhancement of homing properties.138 It is worthwhile noting that this represents a substantial increase above the already efficient rate of homeostatic trafficking whereby ~1 in 4 lymphocytes initiate the adhesive events that precede extravasation.75,129

Fever-range temperatures augment trafficking through a lymphocyte-autonomous mechanism by targeting the binding activity of both L-selectin (FIG. 3a) and α4β7 integrin without altering their density.134,136139 In lymph node HEVs, fever-range hyperthermia promotes L-selectin-dependent lymphocyte rolling along vessel walls through the formation of short-lived catch-bonds with its endothelial counter-receptor, peripheral node addressin (PNAD).74,75 Febrile temperatures also enhance α4β7 integrin binding to mucosal addressin cell adhesion molecule 1 (MADCAM1) in Peyer’s patch and mesenteric lymph node HEVs.144 Direct exposure of lymphocytes to heat does not alter LFA1 affinity for its endothelial ligands.134,136 It remains an open question whether the chemokine receptor, CCR7, is responsive to febrile temperatures.

The intrinsic binding function of HEVs is also enhanced approximately 2-fold in LPS- or turpentine-induced mouse models of fever as well as during exposure of mice to fever-range whole body hyperthermia.136,137,140142 (FIG. 3a). As in lymphocytes, maximal enhancement of HEV adhesion requires sustained temperature elevation (more than 6 hours),136,137,140142 recapitulating the extended time-frame of physiological fever responses. Chen et al. visualized lymphocyte interactions in mouse HEVs using intravital microscopy (FIG. 3b), together with quantitative image analysis of trafficking molecules, to pinpoint the thermally responsive trafficking mechanisms in HEVs.140,141 Thermal stress does not alter the ability of HEVs to support rolling, nor does it change the intraluminal density of the prototypical rolling molecules, PNAD or MADCAM1.140,141 Instead, exposure to febrile temperatures profoundly increases the ability of HEVs to support the stable arrest of lymphocytes and this can be attributed to heat-induced increases in the intravascular density of CCL21 and ICAM1140,141 (FIG. 3a). Of note, the level of HEV adhesiveness and ICAM1 expression induced by thermal stress is equivalent to that observed in response to the potent pro-inflammatory cytokine, TNF.140 Thermal upregulation of CCL21 and ICAM1 in HEVs is consistent with the known concentration-dependent roles of these molecules in augmenting LFA1 affinity (~10,000-fold), thereby supporting stable adhesion of lymphocytes within vessel walls.145,146 Additionally, ICAM1 elevation in response to hyperthermia likely promotes LFA1-dependent transendothelial migration in HEVs and the formation of ICAM1-dense adhesive patches that guide lymphocyte diapedesis into underlying tissues.133,147,148

Once lymphocytes gain entry into lymphoid organs, there is evidence that their ability to respond to stimulatory signals is also enhanced by febrile temperatures. Direct exposure of T cells to fever-range hyperthermia increases their proliferation in response to mitogens.149,150 Furthermore, in both in vitro and in vivo models of antigen-driven T cell activation by APCs, thermally treated CD8+ T cells show greater differentiation towards an effector phenotype, with pronounced L-selectin downregulation, enhanced cytotoxic function and increased production of IFNγ151,152 (FIG. 3a). Enhanced stimulation of naive CD8+ T cells is aligned with temperature-dependent PKCβ activation, prolonged stable contacts with APCs, and transient clustering of components of the immunological synapse (TCRβ and CD8) in cholesterol-enriched microdomains,.151,152 Similar heat-induced changes in membrane fluidity and macromolecular clustering in the plasma membrane occur in CD4+ T cells which reduce the requirement for CD28 stimulation for IL-2 production.153 These findings suggest that febrile temperatures lower the threshold for T cell signalling and effector T cell differentiation by pre-associating the signalling components of the TCR complex.

IL-6 is a thermally sensitive effector of trafficking

Investigation into the mechanisms underlying thermal regulation of trafficking led to the unexpected discovery that the same pyrogenic cytokine responsible for inducing fever, namely IL-6,135,137,138 also controls both lymphocyte and endothelial adhesion.132,138140,142 The thermal response further depends on a second soluble factor, the soluble form of the IL-6 receptor α subunit (sIL-6Rα), which acts cooperatively with IL-6 and the membrane-anchored gp130 signal transducing molecule through a well-defined mechanism termed trans-signalling138140,154,155 (FIG. 4a). This thermally sensitive mechanism was identified in vitro and in vivo using recombinant soluble gp130138,140, which is a competitive antagonist of IL-6 trans-signalling but does not affect classical signalling involving membrane-anchored IL-6Rα.154,156

Figure 4. Thermal stress acts through IL-6 trans-signalling to improve lymphocyte trafficking into lymph nodes.

Figure 4

(a) Heat-dependent interelukin-6 (IL-6) trans-signalling is initiated by binding of the soluble form of the IL-6 receptor α subunit (sIL-6Rα) to both IL-6 and membrane-anchored gp130.154,155 Soluble gp130 functions as a selective antagonist of IL-6 trans-signalling and downstream activation of canonical JAK–STAT and MEK1–ERK1/ERK2 signalling pathways but does not interfere with classical signalling by membrane-anchored IL-6Rα and transmembrane gp130.156 (b) Febrile temperatures act on lymphocytes and high endothelial cells (HECs) to improve lymphocyte trafficking exclusively across high endothelial venules (HEVs) in lymph nodes. Vessel segments immediately proximal to HEVs are refractory to thermal treatment, which may reflect the lower expression of gp130 by non-specialized squamous endothelial cells that line non-HEVs.162 Left inset, fever-range temperatures act directly on lymphocytes through IL-6 trans-signalling to stimulate the MEK1–ERK1/ERK2 signalling pathway, promoting L-selectin adhesion as well as intermolecular interactions between the actin-based cytoskeleton, α-actinin, and the cytoplasmic tail of L-selectin.138 Right inset, IL-6 trans-signalling upregulates the intravascular density of ICAM1 in HEVs during heat treatment of mice, although the downstream signalling mediators remain unknown. Fibroblastic reticular cells that are in direct contact with HECs165 are a possible source of the IL-6 while proximal dendritic cells (DCs) and T cells could provide the sIL-6R138,164 required to enhance the adhesive properties of HEVs during thermal stress.

In lymphocytes, the MEK1–ERK1/ERK2 signalling pathway, but not p38 MAPK or JNK, operates downstream of IL-6–sIL-6Rα trans-signalling in response to heat.138 This promotes L-selectin interactions with actin-based cytoskeletal scaffolding elements, thereby enhancing its apparent tensile strength (FIG. 4b). IL-6-induced activation of STAT3 also occurs in lymphocytes in response to thermal stress138, although it is not known whether this contributes to lymphocyte adhesion, or delivers survival signals157,158 that aid the expansion of populations of effector lymphocytes within lymphoid organs. Consistent with the evolutionary conservation of the febrile response, L-selectin adhesion is induced by fever-range temperatures through a common IL-6 trans-signalling mechanism in animals representing four taxa of jawed vertebrates that includes endothermic mammals (for example, human, rodents, dog, cow, tiger, elephant, and rhinoceros) and birds (chicken), as well as ectothermic amphibians and fish.134,135,137139 These observations strongly suggest that conservation of IL-6-regulated lymphocyte trafficking mechanisms over hundreds of millions of years of evolution confers a survival benefit during fever.

Gp130 ligation by IL-6–sIL-6Rα also upregulates the intravascular density of ICAM1 in HEVs during heat treatment of mice132,140 (FIG. 4b). The dual requirement for IL-6 and sIL-6Rα for ICAM1-dependent trafficking in HEVs during thermal stress is in line with the prevailing view that endothelial cells generally lack membrane-anchored IL-6Rα, and thus are refractory to IL-6 unless sIL-6Rα is available.132 STAT3 and MEK1–ERK1/ERK2 signalling have been implicated in transcriptional regulation of ICAM1132 and, thus, are potential mediators of the thermal response in HEVs. By contrast, CCL21 induction is not dependent on IL-6 trans-signalling,140 suggesting an additional molecular pathway is induced by febrile temperatures.

One of the most intriguing findings to emerge from intravital imaging relates to the tight spatial regulation of IL-6–sIL-6Rα responses in vascular beds during thermal responses. In this regard, Chen et al. showed that HEVs respond to IL-6 trans-signalling during thermal stress, but contiguous vascular segments that are not comprised of high endothelial cells (HECs) are completely refractory to thermally induced IL-6 trans-signalling (FIG. 4b).140 Similarly, non-HEVs in other organs are not responsive to febrile temperatures although heat-shock (which occurs at temperatures greater than 43°C) reportedly stimulates ICAM1 expression in normal vascular endothelium137,140142,159,160 This restricted vascular response to physiological temperature elevation is proposed to maintain focal trafficking of lymphocytes at HEVs in lymph nodes and Peyer’s patches located throughout the body, thus maximizing their opportunity to scan pathogen-derived antigens from peripheral sites of infection.137,140,141

The mechanism maintaining spatial resolution within venular segments over distances spanning the width of a single HEC (~30 µm)161 remains to be resolved, but clues have emerged from recent transcriptional profiling of various cell subsets in lymphoid organs. HECs are distinguished from their normal endothelial cell counterparts by elevated expression of Il6st (which encodes gp130)162 that could theoretically predispose them to be highly sensitive to IL-6–sIL-6Rα in the local milieu (FIG. 4b). Although the overall nodal concentrations of IL-6–sIL-6Rα are unchanged by thermal stress,140,163 heat could theoretically induce their synthesis by discrete cell populations or lower the threshold for signalling in HECs. Fibroblastic reticular cells (FRCs) are a possible source of IL-6 during fever based on their high expression of Il6 mRNA relative to hematopoietic cells or blood endothelial cells within skin-derived lymph nodes.164 Unlike other vascular beds that are circumscribed by pericytes, HEVs are in direct contact with FRCs, and thus are optimally positioned to receive instructions from FRC-derived cytokines.74,75,165 Of particular relevance is a report that IL-6 synthesis by fibroblasts can be induced by the heat-inducible transcription factor, HSF1.166 The sIL-6Rα necessary for trans-signalling is likely provided by neighbouring leukocytes including DCs, monocytes, and/or T cells.138,164 Recent studies have shown that febrile temperatures can also act through IL-6 trans-signalling to augment the recruitment of cytotoxic CD8+ T cells across tumour-associated vessels.142 These studies are highly relevant to the use of thermal medicine as an adjuvant for cancer immunotherapy (BOX 2) and raise the possibility that fever could invoke similar mechanisms to amplify effector T cell trafficking at sites of infection.

BOX 2. Thermal therapy and cancer.

Thermal therapy is administered at a wide range of temperatures for cancer treatment. High temperature focal hyperthermia (> 45°C) and ablation therapy (> 70°C) directly destroy cancer cells and can indirectly boost antitumour immunity, while moderate hyperthermic therapy (38–42°C) is used mainly in an adjuvant setting to target the tumour microenvironment.206208 Temperature effects on blood flow, vascular permeability, interstitial pressure and hypoxia are implicated in enhanced chemo- and radiosensitization in cancer patients treated with hyperthermia.209215 Thermal therapy also holds promise for improving delivery of chemotherapeutic drug cargo by heat-sensitive liposomes.216 Recent preclinical studies suggest that the immunostimulatory activities of febrile temperatures can be exploited therapeutically in combination with promising cancer treatments. Emerging immunotherapies such as dendritic cell (DC) vaccination, adoptive transfer of ex vivo activated T cells, or checkpoint blockade inhibitors (for example, drugs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PD1)) have shown benefit in generating antitumour immunity.217220 Notably, the efficacy of DC vaccines in patients with advanced melanomas or mouse tumour models is substantially improved with the use of hyperthermia as an adjuvant therapy.221,222 Moreover, fever-range thermal therapy overcomes impediments to trafficking in mouse tumour vessels through an interleukin-6 (IL-6) trans-signalling mechanism that stimulates E-selectin and P-selectin-dependent rolling and intracellular adhesion molecule-1 (ICAM1)-dependent firm adhesion of adoptively transferred CD8+ cytotoxic T cells (see figure).142 Increased T cell entry into tumours is further linked to improved antitumour immunity and delayed tumour growth.142 The antitumour immune effects of IL-6 unleashed by thermal therapy are counterintuitive in light of substantial evidence that IL-6 signalling exerts pro-tumourigenic activities by stimulating the survival and proliferation of tumour cells as well as angiogenesis.155,223 Together, these studies highlight a unique role for thermal therapy in modulating the tumour microenvironment that can be co-opted to increase the efficacy of diverse anti-cancer therapies.

BOX 2

A return to homeostasis

The immune response must be tightly regulated to avoid excessive tissue damage after infection. By extension, it makes sense that the effects of febrile temperatures on the immune system are also temporally regulated during the resolution phase of inflammation although a full picture of the underlying mechanisms is yet to emerge. One example is the rapid restoration of lymphocyte trafficking in HEVs to basal levels within 6 hours following cessation of fever-range hyperthermia.134,137,141 Normalization of HEVs is mediated by zinc-dependent metalloproteinases that cleave endothelial ICAM1 while sparing other trafficking molecules (such as PNAD),141 although it is not known if heat stimulates the catalytic activity of these enzymes. In line with a potential anti-inflammatory role of hyperthermic temperatures, heat shock (42°C for 15 min) has been found to blunt leukocyte adhesion within vessels if administered 2 day prior to the intravascular delivery of the neutrophil attractant FMLP in vivo.167

Although febrile temperatures initially increase the production of pro-inflammatory cytokines by macrophages at sites of inflammation,5961,95,96 there is also evidence that thermal stress dampens cytokine synthesis once macrophages become activated. This sequence of events is analgous to natural fever, which often occurs after macrophages and other innate immune cells initially encounter PAMPs. In this regard, human monocyte-derived macrophages with an activated phenotype produce less TNF, IL-6, and IL-1β when exposed to febrile temperatures than heat-inexperienced cells.95,96,168170 Heat reduces transcription of pro-inflammatory cytokines through repressive activities of HSF1, together with diminished recruitment of NF-κB to the promoter regions of cytokine-encoding genes, and also lowers cytokine mRNA stability.171173 Thermal treatment of LPS-activated macrophages also appears to dial down inflammation by inhibiting the release of the inflammatory DAMP known as high mobility group box 1 (HMGB1), which is a ligand for TLR2 and TLR4.170,174 Inhibition of HMGB1 release prevents the subsequent activation of NF-κB, which controls the synthesis of pro-inflammatory cytokines in innate immune cells.169,170,174 The idea that heat can dampen an on-going pro-inflammatory condition in vivo has recently been tested in a murine model of collagen-induced arthritis.175 Mice exposed to fever-range hyperthermia had significantly less joint damage, correlated with a reduction in serum TNF levels and increased IL-10 production in inflamed joints. Collectively, these findings suggest that strategic temperature shifts contribute to a biochemical negative feed-back loop that protects tissues against damage from excessive cytokine release following infection.

Thermogenesis and adrenergic signalling in immunity– an emerging concept

Neural components of the thermoregulatory system continuously monitor temperature changes throughout the body and initiate integrated responses that either increase internal heat content (for example, through thermogenesis in brown adipose tissue) or increase the dissipation of heat (for example, following intense exercise).8 Given the homeostatic importance of thermoregulation, it is all the more remarkable that fever has been so long maintained in evolution, as natural thermoregulatory signals must be suppressed in order to increase body temperature. Although the examples discussed earlier in this review demonstrate that the immune system is responsive to elevated temperatures, new studies reveal that this system is also highly sensitive to the metabolic stress associated with thermogenesis. Emerging evidence strongly supports a direct role for cold stress-induced norepinephrine production and its interaction with β-adrenergic receptors on immune cells as a major mechanism for immune modulation by environmental cold stress. It is well established that norepinephrine-driven stimulation of β-adrenergic receptors is crucial for the release of additional heat from mitochondria in brown adipose tissue during cold stress to maintain a normal core body temperature.176,177 Moreover, the ubiquitous presence of β-adrenergic receptors has been observed on the surface of immune cells, and there is a growing appreciation of the functional consequences of signalling through these receptors.178180 Even more recent studies demonstrate a crucial role for β-adrenergic receptor signalling by norepinephrine for control of lymphocyte egress from lymph nodes and modulation of cytokine production and proliferation in memory CD8+ memory T cells.181,182

These parallel lines of research have now been joined in studies that demonstrate marked alterations in immune cell activity during cold stress. Nguyen et al. discovered that cold stress stimulates IL-4 and IL-13–driven differentiation of macrophages in brown fat toward an ‘alternative activation’ programme leading to their production of norepinephrine.183 (FIG. 5a). Surprisingly, data obtained using various knock-out mice (deficient in IL-4, IL-13, STAT6 or IL-4 receptor) revealed that the norepinephrine produced by these macrophages is critical for maintaining sufficient thermogenesis in the face of cold stress.183,184 Kokolus et al. further demonstrated that DCs exhibit a reduced ability to stimulate T cells if they are from cold-stressed mice that have a normal body temperature due to increased thermogenesis.183,184 Cold stress is also associated with accelerated tumour growth in murine models, which reflects enhanced tumour cell survival pathways as well as a shifted balance toward an immunosuppressive microenvironment with elevated intratumoral myeloid-derived suppressor cells and regulatory T cells together with reduced CD8+ effector T cells184186 (FIG. 5b).

Figure 5. Cold stress stimulates nerve-driven modulation of thermogenesis and anti-tumour immunity.

Figure 5

(a) Exposure to cold stress drives the release of neurotransmitters, such as norepinephrine, by neurons. This initiates the interleukin-4 (IL-4) and IL-13-driven ‘alternative activation’ programme of differentiation in macrophages, resulting in additional production of norepinephrine, which stimulates β-adrenergic receptors (βAR) expressed on brown adipose cells driving thermogenesis.183 (b) Cold stress in tumour-bearing mice maintained at standard housing temperatures (20–26°C) tilts the balance towards an immunosuppressive local tumour microenvironment. This is characterized by a substantial increase in populations of intratumoral myeloid-derived suppressor cells (MDSCs) and regulatory T (Treg) cells and a concomitant decrease in the number of CD8+ T cells when compared to tumours that develop in mice housed under thermoneutral ambient temperature (30–31°C).184 Tumour cell survival and tumour growth are also accelerated by cold stress. 184186

An intriguing aspect is that the presence of cancer creates a notable heat seeking behavioural response in animals.184 These data support the conclusions drawn by Romanovsky and colleagues who have contended that endothermic animals, including humans, exhibit heat-seeking behaviour even before other fever-generating symptoms occur.9,10 Findings in this exciting area contribute additional molecular detail to the fundamental role of temperature stress in influencing the functional balance between various arms of the immune system.187,188

Concluding remarks and future directions

The evolutionary conservation of the fever response over millions of years is in line with its protective role — the survival benefit conferred on the host outweighs the metabolic cost of elevating core body temperatures during infection. Cellular components of the immune system have emerged as central players that actively drive fever induction in addition to serving as thermally sensitive effectors. Moreover, the complexity of the molecular pathways that coordinate a febrile response is mirrored by the diverse cell types that are affacted by hyperthermic temperatures; these include DCs, macrophages, NK cells, neutrophils, T and B lymphocytes, and vascular endothelial cells. The picture that emerges is one in which febrile temperatures serve as a systemic alert system that broadly promotes immune surveillance during challenge by invading pathogens. Furthermore, mechanistic insight into the immune-protective nature of fever has opened new avenues to exploit the immunostimulatory activities of thermal stress in the context of cancer therapy.

Fundamental questions remain regarding the nature of the temperature-sensing machinery that triggers changes in immune cell behaviour. HSF1-regulated HSPs are strong candidates in view of their rapid induction even at the relatively modest temperature elevation (ΔT~1–4°C) that accompanies fever.26,122,189191 Also intriguing are reports that HSF1 regulates additional genes relevant to the induction and/or effector phases of fever, including IL6 and COX2.166,192 Notably, HSP90 and the JAK1–JAK2–STAT3 signalling axis triggered by IL-6 are participants in a feed-forward loop — IL-6-–STAT3 signalling stimulates HSP90 production while JAK2 and STAT3 are established client proteins that are chaperoned by HSP90.193197 Thus, it is tempting to speculate that induction of HSP90 or other HSPs by febrile temperatures lowers the threshold for IL-6 signalling. Additionally, a class of temperature-sensing transient receptor potential (TRP) cation channel proteins expressed on immune cells and endothelial cells are likely to coordinate responses to febrile temperatures and inflammatory cytokines such as IL-6 and lipid mediators.26,198,199 There are also outstanding questions regarding the mechanisms underlying the spatial regulation by IL-6 during fever induction and lymphocyte trafficking in HEVs. Although brain endothelial cells and HECs are predicted to be main targets for IL-6, contributions of intermediary cells have not been excluded.

Another unresolved question is whether febrile temperatures mobilize innate and adaptive immune cells to sites of infection. Observations that administration of fever-range hyperthermia is effective in boosting E-selectin, P-selectin and ICAM1-dependent trafficking of cytotoxic CD8+ T cells in tumour tissues142 raise the strong possibility that similar mechanisms are enacted by fever in infected tissues. Similarly to CXCL8, several inflammatory chemokines that recruit NK cells, CD4+ and CD8+ T cells, and monocytes (CXCL9, CXCL10, CXCL11, CXCL12) contain putative HSF1-binding sites within their gene promoters and, consequently, may be induced by thermal stress.26 An important caveat is that information regarding the cytokine circuitry (for example, IL-6, RANKL and IL-1) leading to fever as well as the impact of temperature on immune function is largely based on experimental models employing LPS or fever-range hyperthermia as surrogates for pathogen-induced fever. Although these studies provide insight into the mechanistic underpinnings for immune regulation by temperatures within the febrile range, lessons learned from studies of thermogenesis183185 indicate that overall temperature sensing (cold or hot) in the absence of disease can have unexpected outcomes on innate and adaptive immunity. The next frontier will be to establish whether the same mechanisms identified during challenge of healthy animals with LPS or fever-range hyperthermia are operative during febrile responses to pathogens.

Acknowledgments

We thank M. Appenheimer, J. Black, and M. Messmer for helpful comments on the manuscript, E. Smith and UC Berkeley Natural Resources Library for assistance with archived citations, and J. Muhitch for providing the photomicrograph depicting lymph node HEV. This work was supported by the US National Institutes of Health (CA79765, CA085183, and AI082039) and the Jennifer Linscott Tietgen Family Foundation. We also acknowledge the significant contributions of colleagues in the field that could not always be cited due to space limitations.

Footnotes

Conflict of interest statement: The authors have no conflicting financial interests.

References

  • 1.Celsus ACA. In: Cornelius Celsus of medicine In eight books. Translated, with notes critical and explanatory, by. Greive James., MD, editor. London: printed for D. Wilson and T. Durham; 1756. [Google Scholar]
  • 2.Kluger MJ. Phylogeny of fever. Fed Proc. 1979;38:30–34. [PubMed] [Google Scholar]
  • 3.Earn DJ, Andrews PW, Bolker BM. Population-level effects of suppressing fever. Proc Biol Sci. 2014;281:20132570. doi: 10.1098/rspb.2013.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Schulman CI, et al. The effect of antipyretic therapy upon outcomes in critically ill patients: a randomized, prospective study. Surg Infect (Larchmt) 2005;6:369–375. doi: 10.1089/sur.2005.6.369. References 3 and 4 show that treatment of fever using anti-pyretic drugs has a detrimental effect on patient outcome during infection.
  • 5.Ryan M, Levy MM. Clinical review: fever in intensive care unit patients. Crit Care. 2003;7:221–225. doi: 10.1186/cc1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kurosawa S, Kobune F, Okuyama K, Sugiura A. Effects of antipyretics in rinderpest virus infection in rabbits. J Infect Dis. 1987;155:991–997. doi: 10.1093/infdis/155.5.991. [DOI] [PubMed] [Google Scholar]
  • 7.Liu E, et al. Naturally occurring hypothermia is more advantageous than fever in severe forms of lipopolysaccharide- and Escherichia coli-induced systemic inflammation. Am J Physiol Regul Integr Comp Physiol. 2012;302:R1372–R1383. doi: 10.1152/ajpregu.00023.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Romanovsky AA. Thermoregulation: some concepts have changed Functional architecture of the thermoregulatory system. Am J Physiol Regul Integr Comp Physiol. 2007;292:R37–R46. doi: 10.1152/ajpregu.00668.2006. [DOI] [PubMed] [Google Scholar]
  • 9.Romanovsky AA, Shido O, Sakurada S, Sugimoto N, Nagasaka T. Endotoxin shock: thermoregulatory mechanisms. Am J Physiol. 1996;270:R693–R703. doi: 10.1152/ajpregu.1996.270.4.R693. [DOI] [PubMed] [Google Scholar]
  • 10.Almeida MC, Steiner AA, Branco LG, Romanovsky AA. Cold-seeking behavior as a thermoregulatory strategy in systemic inflammation. Eur J Neurosci. 2006;23:3359–3367. doi: 10.1111/j.1460-9568.2006.04854.x. [DOI] [PubMed] [Google Scholar]
  • 11.Launey Y, Nesseler N, Malledant Y, Seguin P. Clinical review: fever in septic ICU patients--friend or foe? Crit Care. 2011;15:222. doi: 10.1186/cc10097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Polderman KH. Induced hypothermia and fever control for prevention and treatment of neurological injuries. Lancet. 2008;371:1955–1969. doi: 10.1016/S0140-6736(08)60837-5. [DOI] [PubMed] [Google Scholar]
  • 13.Bernheim HA, Kluger MJ. Fever: effect of drug-induced antipyresis on survival. Science. 1976;193:237–239. doi: 10.1126/science.935867. [DOI] [PubMed] [Google Scholar]
  • 14.Vaughn LK, Bernheim HA, Kluger MJ. Fever in the lizard Dipsosaurus dorsalis. Nature. 1974;252:473–474. doi: 10.1038/252473a0. [DOI] [PubMed] [Google Scholar]
  • 15.Covert JB, Reynolds WW. Survival value of fever in fish. Nature. 1977;267:43–45. doi: 10.1038/267043a0. [DOI] [PubMed] [Google Scholar]
  • 16. Reynolds WW, Casterlin ME, Covert JB. Behavioural fever in teleost fishes. Nature. 1976;259:41–42. doi: 10.1038/259041a0. References 13–16 reveal that ecothermic vertebrates (lizards, fish) develop fever through behavioral changes and that fever is associated with a survival benefit.
  • 17.Simone-Finstrom M, Foo B, Tarpy DR, Starks PT. Impact of Food Availability, Pathogen Exposure, and Genetic Diversity on Thermoregulation in Honey Bees (Apis mellifera) Journal of Insect Behavior. 2014;27:527–539. [Google Scholar]
  • 18.Blanford S, Thomas MB. Adult survival, maturation, and reproduction of the desert locust Schistocerca gregaria infected with the fungus Metarhizium anisopliae var acridum. J Invertebr Pathol. 2001;78:1–8. doi: 10.1006/jipa.2001.5031. [DOI] [PubMed] [Google Scholar]
  • 19.Mackowiak PA. Fever: blessing or curse? A unifying hypothesis. Ann Intern Med. 1994;120:1037–1040. doi: 10.7326/0003-4819-120-12-199406150-00010. [DOI] [PubMed] [Google Scholar]
  • 20.Cabanac M. Fever in the leech, Nephelopsis obscura (Annelida) J Comp Physio B. 1989;159:281–285. [Google Scholar]
  • 21.Yarwood CE. Heat of Respiration of Injured and Diseased Leaves. Phytopathology. 1953;43:675–681. [Google Scholar]
  • 22.Zhu Y, et al. Regulation of thermogenesis in plants: the interaction of alternative oxidase and plant uncoupling mitochondrial protein. J Integr Plant Biol. 2011;53:7–13. doi: 10.1111/j.1744-7909.2010.01004.x. [DOI] [PubMed] [Google Scholar]
  • 23.Pockley AG, Calderwood SK, Santoro MG. Prokaryotic and eukaryotic heat shock proteins in infectious disease. Dordrecht: Springer; 2010. [Google Scholar]
  • 24.Lwoff A. From protozoa to bacteria and virusesFifty years with microbes (Andre Lwoff) Annu Rev Microbiol. 1971;25:1–26. doi: 10.1146/annurev.mi.25.100171.000245. [DOI] [PubMed] [Google Scholar]
  • 25.Osawa E, Muschel LH. Studies Relating to the Serum Resistance of Certain Gram-Negative Bacteria. J Exp Med. 1964;119:41–51. doi: 10.1084/jem.119.1.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hasday JD, Thompson C, Singh IS. Fever, immunity, and molecular adaptations. Compr Physiol. 2014;4:109–148. doi: 10.1002/cphy.c130019. [DOI] [PubMed] [Google Scholar]
  • 27.Morrison SF, Madden CJ, Tupone D. Central control of brown adipose tissue thermogenesis. Front Endocrinol (Lausanne) 2012;3 doi: 10.3389/fendo.2012.00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Taipale M, Jarosz DF, Lindquist S. HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol. 2010;11:515–528. doi: 10.1038/nrm2918. [DOI] [PubMed] [Google Scholar]
  • 29.Akerfelt M, Morimoto RI, Sistonen L. Heat shock factors: integrators of cell stress, development and lifespan. Nat Rev Mol Cell Biol. 2010;11:545–555. doi: 10.1038/nrm2938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Saper CB, Romanovsky AA, Scammell TE. Neural circuitry engaged by prostaglandins during the sickness syndrome. Nat Neurosci. 2012;15:1088–1095. doi: 10.1038/nn.3159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Matsumura K, et al. Brain endothelial cells express cyclooxygenase-2 during lipopolysaccharide-induced fever: light and electron microscopic immunocytochemical studies. J Neurosci. 1998;18:6279–6289. doi: 10.1523/JNEUROSCI.18-16-06279.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yamagata K, et al. Coexpression of microsomal-type prostaglandin E synthase with cyclooxygenase-2 in brain endothelial cells of rats during endotoxin-induced fever. J Neurosci. 2001;21:2669–2677. doi: 10.1523/JNEUROSCI.21-08-02669.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Engstrom L, et al. Lipopolysaccharide-induced fever depends on prostaglandin E2 production specifically in brain endothelial cells. Endocrinology. 2012;153:4849–4861. doi: 10.1210/en.2012-1375. [DOI] [PubMed] [Google Scholar]
  • 34.Blatteis CM, Li S, Li Z, Feleder C, Perlik V. Cytokines, PGE2 and endotoxic fever: a re-assessment. Prostaglandins Other Lipid Mediat. 2005;76:1–18. doi: 10.1016/j.prostaglandins.2005.01.001. [DOI] [PubMed] [Google Scholar]
  • 35. Steiner AA, Chakravarty S, Rudaya AY, Herkenham M, Romanovsky AA. Bacterial lipopolysaccharide fever is initiated via Toll-like receptor 4 on hematopoietic cells. Blood. 2006;107:4000–4002. doi: 10.1182/blood-2005-11-4743. References 34–35 show that expression and release of prostaglandin E2 from hematopoetic cells is required for the development of fever during LPS challenge.
  • 36.Roth J, Blatteis CM. Mechanisms of fever production and lysis: lessons from experimental LPS fever. Compr Physiol. 2014;4:1563–1604. doi: 10.1002/cphy.c130033. [DOI] [PubMed] [Google Scholar]
  • 37.Ivanov AI, Romanovsky AA. Prostaglandin E2 as a mediator of fever: synthesis and catabolism. Front Biosci. 2004;9:1977–1993. doi: 10.2741/1383. [DOI] [PubMed] [Google Scholar]
  • 38.Furuyashiki T, Narumiya S. Stress responses: the contribution of prostaglandin E(2) and its receptors. Nat Rev Endocrinol. 2011;7:163–175. doi: 10.1038/nrendo.2010.194. [DOI] [PubMed] [Google Scholar]
  • 39.Engblom D, et al. Microsomal prostaglandin E synthase-1 is the central switch during immune-induced pyresis. Nat Neurosci. 2003;6:1137–1138. doi: 10.1038/nn1137. [DOI] [PubMed] [Google Scholar]
  • 40. Lazarus M, et al. EP3 prostaglandin receptors in the median preoptic nucleus are critical for fever responses. Nat Neurosci. 2007;10:1131–1133. doi: 10.1038/nn1949. References 39–40 demonstrate that responses via EP3 prostaglandin receptors is required for the development of fever.
  • 41.Nakamura K, Morrison SF. A thermosensory pathway that controls body temperature. Nat Neurosci. 2008;11:62–71. doi: 10.1038/nn2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dantzer R, Wollman E. Molecular mechanisms of fever: the missing links. Eur Cytokine Netw. 1998;9:27–31. [PubMed] [Google Scholar]
  • 43.Steinman L. Nuanced roles of cytokines in three major human brain disorders. J Clin Invest. 2008;118:3557–3563. doi: 10.1172/JCI36532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 45.O'Neill LA, Golenbock D, Bowie AG. The history of Toll-like receptors - redefining innate immunity. Nat Rev Immunol. 2013;13:453–460. doi: 10.1038/nri3446. [DOI] [PubMed] [Google Scholar]
  • 46.Elander L, Ruud J, Korotkova M, Jakobsson PJ, Blomqvist A. Cyclooxygenase-1 mediates the immediate corticosterone response to peripheral immune challenge induced by lipopolysaccharide. Neurosci Lett. 2010;470:10–12. doi: 10.1016/j.neulet.2009.12.036. [DOI] [PubMed] [Google Scholar]
  • 47. Hanada R, et al. Central control of fever and female body temperature by RANKL/RANK. Nature. 2009;462:505–509. doi: 10.1038/nature08596. This study identifies a pivotal role for RANKL in the brain in generating a fever in response to LPS.
  • 48.Benveniste EN, Sparacio SM, Norris JG, Grenett HE, Fuller GM. Induction and regulation of interleukin-6 gene expression in rat astrocytes. J Neuroimmunol. 1990;30:201–212. doi: 10.1016/0165-5728(90)90104-u. [DOI] [PubMed] [Google Scholar]
  • 49.Beurel E, Jope RS. Lipopolysaccharide-induced interleukin-6 production is controlled by glycogen synthase kinase-3 and STAT3 in the brain. J Neuroinflammation. 2009;6:9. doi: 10.1186/1742-2094-6-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sawada M, Suzumura A, Marunouchi T. TNF alpha induces IL-6 production by astrocytes but not by microglia. Brain Res. 1992;583:296–299. doi: 10.1016/s0006-8993(10)80037-x. [DOI] [PubMed] [Google Scholar]
  • 51.Woodroofe MN, et al. Detection of interleukin-1 and interleukin-6 in adult rat brain, following mechanical injury, by in vivo microdialysis: evidence of a role for microglia in cytokine production. J Neuroimmunol. 1991;33:227–236. doi: 10.1016/0165-5728(91)90110-s. [DOI] [PubMed] [Google Scholar]
  • 52.Ringheim GE, Burgher KL, Heroux JA. Interleukin-6 mRNA expression by cortical neurons in culture: evidence for neuronal sources of interleukin-6 production in the brain. J Neuroimmunol. 1995;63:113–123. doi: 10.1016/0165-5728(95)00134-4. [DOI] [PubMed] [Google Scholar]
  • 53.Vallieres L, Rivest S. Regulation of the genes encoding interleukin-6, its receptor, and gp130 in the rat brain in response to the immune activator lipopolysaccharide and the proinflammatory cytokine interleukin-1beta. J Neurochem. 1997;69:1668–1683. doi: 10.1046/j.1471-4159.1997.69041668.x. [DOI] [PubMed] [Google Scholar]
  • 54.Cartmell T, Poole S, Turnbull AV, Rothwell NJ, Luheshi GN. Circulating interleukin-6 mediates the febrile response to localised inflammation in rats. J Physiol. 2000;(526 Pt 3):653–661. doi: 10.1111/j.1469-7793.2000.00653.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chai Z, Gatti S, Toniatti C, Poli V, Bartfai T. Interleukin (IL)-6 gene expression in the central nervous system is necessary for fever response to lipopolysaccharide or IL-1 beta: a study on IL-6-deficient mice. J Exp Med. 1996;183:311–316. doi: 10.1084/jem.183.1.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kozak W, et al. IL-6 IL-1 beta in fever Studies using cytokine-deficient (knockout) mice. Ann N Y Acad Sci. 1998;856:33–47. doi: 10.1111/j.1749-6632.1998.tb08310.x. [DOI] [PubMed] [Google Scholar]
  • 57. Hamzic N, et al. Interleukin-6 primarily produced by non-hematopoietic cells mediates the lipopolysaccharide-induced febrile response. Brain Behav Immun. 2013;33:123–130. doi: 10.1016/j.bbi.2013.06.006. References 55–57 highlight that loss of IL-6 signaling alone abrogates fever in response in LPS or IL-1-induced inflammatory models.
  • 58.Nilsberth C, Hamzic N, Norell M, Blomqvist A. Peripheral lipopolysaccharide administration induces cytokine mRNA expression in the viscera and brain of fever-refractory mice lacking microsomal prostaglandin E synthase-1. J Neuroendocrinol. 2009;21:715–721. doi: 10.1111/j.1365-2826.2009.01888.x. [DOI] [PubMed] [Google Scholar]
  • 59.Jiang Q, et al. Exposure to febrile temperature upregulates expression of pyrogenic cytokines in endotoxin-challenged mice. Am J Physiol. 1999;276:R1653–R1660. doi: 10.1152/ajpregu.1999.276.6.R1653. [DOI] [PubMed] [Google Scholar]
  • 60.Ostberg JR, Taylor SL, Baumann H, Repasky EA. Regulatory effects of fever-range whole-body hyperthermia on the LPS-induced acute inflammatory response. J Leukoc Biol. 2000;68:815–820. [PubMed] [Google Scholar]
  • 61. Rice P, et al. Febrile-range hyperthermia augments neutrophil accumulation and enhances lung injury in experimental gram-negative bacterial pneumonia. J Immunol. 2005;174:3676–3685. doi: 10.4049/jimmunol.174.6.3676. This report establishes that febrile temperatures drive IL-8 expression to initiate neutrophil infiltration in the lungs.
  • 62.Grupp SA, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Teachey DT, et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood. 2013;121:5154–5157. doi: 10.1182/blood-2013-02-485623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cao C, Matsumura K, Yamagata K, Watanabe Y. Endothelial cells of the rat brain vasculature express cyclooxygenase-2 mRNA in response to systemic interleukin-1 beta: a possible site of prostaglandin synthesis responsible for fever. Brain Res. 1996;733:263–272. doi: 10.1016/0006-8993(96)00575-6. [DOI] [PubMed] [Google Scholar]
  • 65.Konsman JP, Vigues S, Mackerlova L, Bristow A, Blomqvist A. Rat brain vascular distribution of interleukin-1 type-1 receptor immunoreactivity: relationship to patterns of inducible cyclooxygenase expression by peripheral inflammatory stimuli. J Comp Neurol. 2004;472:113–129. doi: 10.1002/cne.20052. [DOI] [PubMed] [Google Scholar]
  • 66.Rummel C, Sachot C, Poole S, Luheshi GN. Circulating interleukin-6 induces fever through a STAT3-linked activation of COX-2 in the brain. Am J Physiol Regul Integr Comp Physiol. 2006;291:R1316–R1326. doi: 10.1152/ajpregu.00301.2006. [DOI] [PubMed] [Google Scholar]
  • 67.Turnbull AV, Prehar S, Kennedy AR, Little RA, Hopkins SJ. Interleukin-6 is an afferent signal to the hypothalamo-pituitary-adrenal axis during local inflammation in mice. Endocrinology. 2003;144:1894–1906. doi: 10.1210/en.2002-220964. [DOI] [PubMed] [Google Scholar]
  • 68.Sundgren-Andersson AK, Ostlund P, Bartfai T. IL-6 is essential in TNF-alpha-induced fever. Am J Physiol. 1998;275:R2028–R2034. doi: 10.1152/ajpregu.1998.275.6.R2028. [DOI] [PubMed] [Google Scholar]
  • 69.Li S, Goorha S, Ballou LR, Blatteis CM. Intracerebroventricular interleukin-6, macrophage inflammatory protein-1 beta and IL-18: pyrogenic and PGE(2)-mediated? Brain Res. 2003;992:76–84. doi: 10.1016/j.brainres.2003.08.033. [DOI] [PubMed] [Google Scholar]
  • 70.Nilsberth C, et al. The role of interleukin-6 in lipopolysaccharide-induced fever by mechanisms independent of prostaglandin E2. Endocrinology. 2009;150:1850–1860. doi: 10.1210/en.2008-0806. [DOI] [PubMed] [Google Scholar]
  • 71.Kong YY, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397:315–323. doi: 10.1038/16852. [DOI] [PubMed] [Google Scholar]
  • 72.Hashizume M, Mihara M. The roles of interleukin-6 in the pathogenesis of rheumatoid arthritis. Arthritis. 2011;2011:765624. doi: 10.1155/2011/765624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Soehnlein O, Lindbom L. Phagocyte partnership during the onset and resolution of inflammation. Nat Rev Immunol. 2010;10:427–439. doi: 10.1038/nri2779. [DOI] [PubMed] [Google Scholar]
  • 74.Girard JP, Moussion C, Forster R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol. 2012;12:762–773. doi: 10.1038/nri3298. [DOI] [PubMed] [Google Scholar]
  • 75.von Andrian UH, Mempel TR. Homing and cellular traffic in lymph nodes. Nat Rev Immunol. 2003;3:867–878. doi: 10.1038/nri1222. [DOI] [PubMed] [Google Scholar]
  • 76.Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659–702. doi: 10.1146/annurev-immunol-032713-120145. [DOI] [PubMed] [Google Scholar]
  • 77.Ostberg JR, Ertel BR, Lanphere JA. An important role for granulocytes in the thermal regulation of colon tumor growth. Immunol Invest. 2005;34:259–272. doi: 10.1081/imm-200064477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Takada Y, et al. Granulocyte-colony stimulating factor enhances anti-tumour effect of hyperthermia. Int J Hyperthermia. 2000;16:275–286. doi: 10.1080/026567300285286. [DOI] [PubMed] [Google Scholar]
  • 79.Bernheim HA, Bodel PT, Askenase PW, Atkins E. Effects of fever on host defense mechanisms after infection in the lizard Dipsosaurus dorsalis. Br J Exp Pathol. 1978;59:76–84. [PMC free article] [PubMed] [Google Scholar]
  • 80.Ellis GS, et al. G-CSF, but not corticosterone, mediates circulating neutrophilia induced by febrile-range hyperthermia. J Appl Physiol (1985) 2005;98:1799–1804. doi: 10.1152/japplphysiol.01376.2004. [DOI] [PubMed] [Google Scholar]
  • 81.Hasday JD, et al. Febrile-range hyperthermia augments pulmonary neutrophil recruitment and amplifies pulmonary oxygen toxicity. Am J Pathol. 2003;162:2005–2017. doi: 10.1016/S0002-9440(10)64333-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Capitano ML, et al. Elevating body temperature enhances hematopoiesis and neutrophil recovery after total body irradiation in an IL-1-, IL-17-, and G-CSF-dependent manner. Blood. 2012;120:2600–2609. doi: 10.1182/blood-2012-02-409805. This study demonstrates that fever-range heating promotes neutrophil release from the bone marrow and is protective following irradiation-induced neutropenia.
  • 83.Ostberg JR, Repasky EA. Comparison of the effects of two different whole body hyperthermia protocols on the distribution of murine leukocyte populations. Int J Hyperthermia. 2000;16:29–43. doi: 10.1080/026567300285402. [DOI] [PubMed] [Google Scholar]
  • 84.Tulapurkar ME, et al. Febrile-range hyperthermia modifies endothelial and neutrophilic functions to promote extravasation. Am J Respir Cell Mol Biol. 2012;46:807–814. doi: 10.1165/rcmb.2011-0378OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Singh IS, et al. Heat shock co-activates interleukin-8 transcription. Am J Respir Cell Mol Biol. 2008;39:235–242. doi: 10.1165/rcmb.2007-0294OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zanker KS, Lange J. Whole body hyperthermia and natural killer cell activity. Lancet. 1982;1:1079–1080. doi: 10.1016/s0140-6736(82)92142-0. [DOI] [PubMed] [Google Scholar]
  • 87.Shen RN, Hornback NB, Shidnia H, Shupe RE, Brahmi Z. Whole-body hyperthermia decreases lung metastases in lung tumor-bearing mice, possibly via a mechanism involving natural killer cells. J Clin Immunol. 1987;7:246–253. doi: 10.1007/BF00915730. [DOI] [PubMed] [Google Scholar]
  • 88.Burd R, et al. Tumor cell apoptosis, lymphocyte recruitment and tumor vascular changes are induced by low temperature, long duration (fever-like) whole body hyperthermia. J Cell Physiol. 1998;177:137–147. doi: 10.1002/(SICI)1097-4652(199810)177:1<137::AID-JCP15>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  • 89.Kappel M, Stadeager C, Tvede N, Galbo H, Pedersen BK. Effects of in vivo hyperthermia on natural killer cell activity, in vitro proliferative responses and blood mononuclear cell subpopulations. Clin Exp Immunol. 1991;84:175–180. doi: 10.1111/j.1365-2249.1991.tb08144.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J Leukoc Biol. 2007;82:1322–1331. doi: 10.1189/jlb.1106699. [DOI] [PubMed] [Google Scholar]
  • 91.Multhoff G, Botzler C, Wiesnet M, Eissner G, Issels R. CD3- large granular lymphocytes recognize a heat-inducible immunogenic determinant associated with the 72-kD heat shock protein on human sarcoma cells. Blood. 1995;86:1374–1382. [PubMed] [Google Scholar]
  • 92.Shi H, et al. Hyperthermia enhances CTL cross-priming. J Immunol. 2006;176:2134–2141. doi: 10.4049/jimmunol.176.4.2134. [DOI] [PubMed] [Google Scholar]
  • 93.Jiang Q, et al. Febrile core temperature is essential for optimal host defense in bacterial peritonitis. Infect Immun. 2000;68:1265–1270. doi: 10.1128/iai.68.3.1265-1270.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hasday JD, Fairchild KD, Shanholtz C. The role of fever in the infected host. Microbes Infect. 2000;2:1891–1904. doi: 10.1016/s1286-4579(00)01337-x. [DOI] [PubMed] [Google Scholar]
  • 95.Lee CT, Zhong L, Mace TA, Repasky EA. Elevation in body temperature to fever range enhances and prolongs subsequent responsiveness of macrophages to endotoxin challenge. PLoS One. 2012;7:e30077. doi: 10.1371/journal.pone.0030077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee CT, Repasky EA. Opposing roles for heat and heat shock proteins in macrophage functions during inflammation: a function of cell activation state? Front Immunol. 2012;3:140. doi: 10.3389/fimmu.2012.00140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Pritchard MT, Li Z, Repasky EA. Nitric oxide production is regulated by fever-range thermal stimulation of murine macrophages. J Leukoc Biol. 2005;78:630–638. doi: 10.1189/jlb.0404220. [DOI] [PubMed] [Google Scholar]
  • 98.Gupta A, et al. Toll-like receptor agonists and febrile range hyperthermia synergize to induce heat shock protein 70 expression and extracellular release. J Biol Chem. 2013;288:2756–2766. doi: 10.1074/jbc.M112.427336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Multhoff G. Heat shock protein 70 (Hsp70): membrane location, export and immunological relevance. Methods. 2007;43:229–237. doi: 10.1016/j.ymeth.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 100.Vega VL, et al. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J Immunol. 2008;180:4299–4307. doi: 10.4049/jimmunol.180.6.4299. [DOI] [PubMed] [Google Scholar]
  • 101.Noessner E, et al. Tumor-derived heat shock protein 70 peptide complexes are cross-presented by human dendritic cells. J Immunol. 2002;169:5424–5432. doi: 10.4049/jimmunol.169.10.5424. [DOI] [PubMed] [Google Scholar]
  • 102.Basu S, Binder RJ, Ramalingam T, Srivastava PK. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity. 2001;14:303–313. doi: 10.1016/s1074-7613(01)00111-x. [DOI] [PubMed] [Google Scholar]
  • 103.Binder RJ, Srivastava PK. Essential role of CD91 in re-presentation of gp96-chaperoned peptides. Proc Natl Acad Sci U S A. 2004;101:6128–6133. doi: 10.1073/pnas.0308180101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Berwin B, et al. Scavenger receptor-A mediates gp96/GRP94 and calreticulin internalization by antigen-presenting cells. EMBO J. 2003;22:6127–6136. doi: 10.1093/emboj/cdg572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Becker T, Hartl FU, Wieland F. CD40, an extracellular receptor for binding and uptake of Hsp70-peptide complexes. J Cell Biol. 2002;158:1277–1285. doi: 10.1083/jcb.200208083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Arnouk H, et al. Tumour secreted grp170 chaperones full-length protein substrates and induces an adaptive anti-tumour immune response in vivo. Int J Hyperthermia. 2010;26:366–375. doi: 10.3109/02656730903485910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Asea A, et al. Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002;277:15028–15034. doi: 10.1074/jbc.M200497200. [DOI] [PubMed] [Google Scholar]
  • 108.Facciponte JG, Wang XY, Subjeck JR. Hsp110 and Grp170, members of the Hsp70 superfamily, bind to scavenger receptor-A and scavenger receptor expressed by endothelial cells-I. Eur J Immunol. 2007;37:2268–2279. doi: 10.1002/eji.200737127. [DOI] [PubMed] [Google Scholar]
  • 109.Lehner T, et al. Heat shock proteins generate beta-chemokines which function as innate adjuvants enhancing adaptive immunity. Eur J Immunol. 2000;30:594–603. doi: 10.1002/1521-4141(200002)30:2<594::AID-IMMU594>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 110.Panjwani NN, Popova L, Srivastava PK. Heat shock proteins gp96 and hsp70 activate the release of nitric oxide by APCs. J Immunol. 2002;168:2997–3003. doi: 10.4049/jimmunol.168.6.2997. [DOI] [PubMed] [Google Scholar]
  • 111.Snyder YM, Guthrie L, Evans GF, Zuckerman SH. Transcriptional inhibition of endotoxin-induced monokine synthesis following heat shock in murine peritoneal macrophages. J Leukoc Biol. 1992;51:181–187. doi: 10.1002/jlb.51.2.181. [DOI] [PubMed] [Google Scholar]
  • 112.Yenari MA, et al. Antiapoptotic and anti-inflammatory mechanisms of heat-shock protein protection. Ann N Y Acad Sci. 2005;1053:74–83. doi: 10.1196/annals.1344.007. [DOI] [PubMed] [Google Scholar]
  • 113.Chen H, et al. Hsp70 inhibits lipopolysaccharide-induced NF-kappaB activation by interacting with TRAF6 and inhibiting its ubiquitination. FEBS Lett. 2006;580:3145–3152. doi: 10.1016/j.febslet.2006.04.066. [DOI] [PubMed] [Google Scholar]
  • 114.Schmitt E, Gehrmann M, Brunet M, Multhoff G, Garrido C. Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy. J Leukoc Biol. 2007;81:15–27. doi: 10.1189/jlb.0306167. [DOI] [PubMed] [Google Scholar]
  • 115.Manzella JP, Roberts NJ., Jr Human macrophage and lymphocyte responses to mitogen stimulation after exposure to influenza virus, ascorbic acid, and hyperthermia. J Immunol. 1979;123:1940–1944. [PubMed] [Google Scholar]
  • 116.Postic B, DeAngelis C, Breinig MK, Monto HO. Effect of temperature on the induction of interferons by endotoxin and virus. J Bacteriol. 1966;91:1277–1281. doi: 10.1128/jb.91.3.1277-1281.1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Knippertz I, et al. Mild hyperthermia enhances human monocyte-derived dendritic cell functions and offers potential for applications in vaccination strategies. Int J Hyperthermia. 2011;27:591–603. doi: 10.3109/02656736.2011.589234. [DOI] [PubMed] [Google Scholar]
  • 118.van Bruggen I, Robertson TA, Papadimitriou JM. The effect of mild hyperthermia on the morphology and function of murine resident peritoneal macrophages. Exp Mol Pathol. 1991;55:119–134. doi: 10.1016/0014-4800(91)90047-2. [DOI] [PubMed] [Google Scholar]
  • 119.Bachleitner-Hofmann T, et al. Heat shock treatment of tumor lysate-pulsed dendritic cells enhances their capacity to elicit antitumor T cell responses against medullary thyroid carcinoma. J Clin Endocrinol Metab. 2006;91:4571–4577. doi: 10.1210/jc.2006-0971. [DOI] [PubMed] [Google Scholar]
  • 120.Yan X, et al. Fever range temperature promotes TLR4 expression and signaling in dendritic cells. Life Sci. 2007;80:307–313. doi: 10.1016/j.lfs.2006.09.022. [DOI] [PubMed] [Google Scholar]
  • 121. Hatzfeld-Charbonnier AS, et al. Influence of heat stress on human monocyte-derived dendritic cell functions with immunotherapeutic potential for antitumor vaccines. J Leukoc Biol. 2007;81:1179–1187. doi: 10.1189/jlb.0506347. This reference shows that exposure of monocyte-derived human dendritic cells to febrile temperatures improves their migratory response to chemokines in vitro and their ability to stimulate naive T cell activation.
  • 122.Ostberg JR, Kaplan KC, Repasky EA. Induction of stress proteins in a panel of mouse tissues by fever-range whole body hyperthermia. Int J Hyperthermia. 2002;18:552–562. doi: 10.1080/02656730210166168. [DOI] [PubMed] [Google Scholar]
  • 123.Peng JC, et al. Monocyte-derived DC primed with TLR agonists secrete IL-12p70 in a CD40-dependent manner under hyperthermic conditions. J Immunother. 2006;29:606–615. doi: 10.1097/01.cji.0000211308.82997.4e. [DOI] [PubMed] [Google Scholar]
  • 124.Ostberg JR, Gellin C, Patel R, Repasky EA. Regulatory potential of fever-range whole body hyperthermia on Langerhans cells and lymphocytes in an antigen-dependent cellular immune response. J Immunol. 2001;167:2666–2670. doi: 10.4049/jimmunol.167.5.2666. [DOI] [PubMed] [Google Scholar]
  • 125.Tal O, et al. DC mobilization from the skin requires docking to immobilized CCL21 on lymphatic endothelium and intralymphatic crawling. J Exp Med. 2011;208:2141–2153. doi: 10.1084/jem.20102392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Schumann K, et al. Immobilized chemokine fields and soluble chemokine gradients cooperatively shape migration patterns of dendritic cells. Immunity. 2010;32:703–713. doi: 10.1016/j.immuni.2010.04.017. [DOI] [PubMed] [Google Scholar]
  • 127.Weber M, et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science. 2013;339:328–332. doi: 10.1126/science.1228456. [DOI] [PubMed] [Google Scholar]
  • 128. Ulvmar MH, et al. The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes. Nat Immunol. 2014;15:623–630. doi: 10.1038/ni.2889. References 125–128 describe the role of CCR7 in sensing CCL21 gradients that direct the migration of mature dendritic cells into the afferent lymphatics and within draining lymph nodes.
  • 129.von Andrian UH. Intravital microscopy of the peripheral lymph node microcirculation in mice. Microcirculation. 1996;3:287–300. doi: 10.3109/10739689609148303. [DOI] [PubMed] [Google Scholar]
  • 130.Blattman JN, et al. Estimating the precursor frequency of naive antigen-specific CD8 T cells. J Exp Med. 2002;195:657–664. doi: 10.1084/jem.20001021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pabst R, Binns RM. Heterogeneity of lymphocyte homing physiology: several mechanisms operate in the control of migration to lymphoid and non-lymphoid organs in vivo. Immunol Rev. 1989;108:83–109. doi: 10.1111/j.1600-065x.1989.tb00014.x. [DOI] [PubMed] [Google Scholar]
  • 132.Vardam TD, et al. Regulation of a lymphocyte-endothelial-IL-6 trans-signaling axis by fever-range thermal stress: hot spot of immune surveillance. Cytokine. 2007;39:84–96. doi: 10.1016/j.cyto.2007.07.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Boscacci RT, et al. Comprehensive analysis of lymph node stroma-expressed Ig superfamily members reveals redundant and nonredundant roles for ICAM-1, ICAM-2, and VCAM-1 in lymphocyte homing. Blood. 2010;116:915–925. doi: 10.1182/blood-2009-11-254334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Wang WC, et al. Fever-range hyperthermia enhances L-selectin-dependent adhesion of lymphocytes to vascular endothelium. J Immunol. 1998;160:961–969. [PubMed] [Google Scholar]
  • 135.Evans SS, et al. Dynamic association of L-selectin with the lymphocyte cytoskeletal matrix. J Immunol. 1999;162:3615–3624. [PubMed] [Google Scholar]
  • 136.Evans SS, Bain MD, Wang WC. Fever-range hyperthermia stimulates alpha4beta7 integrin-dependent lymphocyte-endothelial adhesion. Int J Hyperthermia. 2000;16:45–59. doi: 10.1080/026567300285411. [DOI] [PubMed] [Google Scholar]
  • 137.Evans SS, et al. Fever-range hyperthermia dynamically regulates lymphocyte delivery to high endothelial venules. Blood. 2001;97:2727–2733. doi: 10.1182/blood.v97.9.2727. [DOI] [PubMed] [Google Scholar]
  • 138.Chen Q, et al. Central role of IL-6 receptor signal-transducing chain gp130 in activation of L-selectin adhesion by fever-range thermal stress. Immunity. 2004;20:59–70. doi: 10.1016/s1074-7613(03)00358-3. [DOI] [PubMed] [Google Scholar]
  • 139. Appenheimer MM, et al. Conservation of IL-6 trans-signaling mechanisms controlling L-selectin adhesion by fever-range thermal stress. Eur J Immunol. 2007;37:2856–2867. doi: 10.1002/eji.200636421. References 138–139 identify an evolutionarily conserved role of IL-6 transignaling in enhancing L-selectin dependent adhesion in lymphocytes during exposure to febrile temperatures.
  • 140. Chen Q, et al. Fever-range thermal stress promotes lymphocyte trafficking across high endothelial venules via an interleukin 6 trans-signaling mechanism. Nat Immunol. 2006;7:1299–1308. doi: 10.1038/ni1406. This study establishes that fever-range thermal stress acts through an IL-6 trans-signaling dependent mechanism to upregulate ICAM1 expression selectively in HEVs.
  • 141. Chen Q, et al. Thermal facilitation of lymphocyte trafficking involves temporal induction of intravascular ICAM-1. Microcirculation. 2009;16:143–158. doi: 10.1080/10739680802353850. This study reveals the restricted temporal nature of ICAM1 induction on HEV in response to fever-range hyperthermia and that restoration of normal trafficking involves a zinc-dependent metalloproteinase-dependent mechanism.
  • 142. Fisher DT, et al. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest. 2011;121:3846–3859. doi: 10.1172/JCI44952. This study demonstrates that febrile-range thermal therapy acts through IL-6 trans-signaling to promote the trafficking of CD8+ effector T cells into the tumor microenvironment and delays tumor growth.
  • 143.Kraybill WG, et al. A phase I study of fever-range whole body hyperthermia (FR-WBH) in patients with advanced solid tumours: correlation with mouse models. Int J Hyperthermia. 2002;18:253–266. doi: 10.1080/02656730110116704. [DOI] [PubMed] [Google Scholar]
  • 144.Picker LJ, Butcher EC. Physiological and molecular mechanisms of lymphocyte homing. Annu Rev Immunol. 1992;10:561–591. doi: 10.1146/annurev.iy.10.040192.003021. [DOI] [PubMed] [Google Scholar]
  • 145.Carman CV, Springer T. AIntegrin avidity regulation: are changes in affinity and conformation underemphasized? Curr Opin Cell Biol. 2003;15:547–556. doi: 10.1016/j.ceb.2003.08.003. [DOI] [PubMed] [Google Scholar]
  • 146.Schurpf T, Springer TA. Regulation of integrin affinity on cell surfaces. EMBO J. 2011;30:4712–4727. doi: 10.1038/emboj.2011.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Carman CV, Springer TA. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol. 2004;167:377–388. doi: 10.1083/jcb.200404129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Carman CV, Springer TA. Trans-cellular migration: cell-cell contacts get intimate. Curr Opin Cell Biol. 2008;20:533–540. doi: 10.1016/j.ceb.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Roberts NJ, Jr, Steigbigel RT. Hyperthermia and human leukocyte functions: effects on response of lymphocytes to mitogen and antigen and bactericidal capacity of monocytes and neutrophils. Infect Immun. 1977;18:673–679. doi: 10.1128/iai.18.3.673-679.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Smith JB, Knowlton RP, Agarwal SS. Human lymphocyte responses are enhanced by culture at 40 degrees C. J Immunol. 1978;121:691–694. [PubMed] [Google Scholar]
  • 151. Mace TA, et al. Differentiation of CD8+ T cells into effector cells is enhanced by physiological range hyperthermia. J Leukoc Biol. 2011;90:951–962. doi: 10.1189/jlb.0511229. This study shows that fever-range temperatures improve T cell activation by altering the fluidity of the plasma membrane and by pre-association of the signalling components of the TCR complex.
  • 152.Mace TA, Zhong L, Kokolus KM, Repasky EA. Effector CD8+ T cell IFN-gamma production and cytotoxicity are enhanced by mild hyperthermia. Int J Hyperthermia. 2012;28:9–18. doi: 10.3109/02656736.2011.616182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zynda E, et al. A role for the thermal microenvironment in co-stimulation requirements during T cell activation. Cell Cycle. 2015 doi: 10.1080/15384101.2015.1049782. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Calabrese LH, Rose-John S. IL-6 biology: implications for clinical targeting in rheumatic disease. Nat Rev Rheumatol. 2014 doi: 10.1038/nrrheum.2014.127. [DOI] [PubMed] [Google Scholar]
  • 155.Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol. 2014;26:38–47. doi: 10.1016/j.smim.2014.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Jostock T, et al. Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem. 2001;268:160–167. doi: 10.1046/j.1432-1327.2001.01867.x. [DOI] [PubMed] [Google Scholar]
  • 157.Atreya R, et al. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat Med. 2000;6:583–588. doi: 10.1038/75068. [DOI] [PubMed] [Google Scholar]
  • 158.Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809. doi: 10.1038/nrc2734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Shah A, et al. Cytokine and adhesion molecule expression in primary human endothelial cells stimulated with fever-range hyperthermia. Int J Hyperthermia. 2002;18:534–551. doi: 10.1080/02656730210157843. [DOI] [PubMed] [Google Scholar]
  • 160.Lefor AT, et al. Hyperthermia increases intercellular adhesion molecule-1 expression and lymphocyte adhesion to endothelial cells. Surgery. 1994;116:214–220. discussion 220–1. [PubMed] [Google Scholar]
  • 161.Ager A. Isolation and culture of high endothelial cells from rat lymph nodes. J Cell Sci. 1987;87(Pt 1):133–144. doi: 10.1242/jcs.87.1.133. [DOI] [PubMed] [Google Scholar]
  • 162.Lee M, et al. Transcriptional programs of lymphoid tissue capillary and high endothelium reveal control mechanisms for lymphocyte homing. Nat Immunol. 2014;15:982–995. doi: 10.1038/ni.2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Mikucki ME, et al. Preconditioning thermal therapy: flipping the switch on IL-6 for anti-tumour immunity. Int J Hyperthermia. 2013;29:464–473. doi: 10.3109/02656736.2013.807440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Malhotra D, et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat Immunol. 2012;13:499–510. doi: 10.1038/ni.2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Katakai T, Hara T, Sugai M, Gonda H, Shimizu A. Lymph node fibroblastic reticular cells construct the stromal reticulum via contact with lymphocytes. J Exp Med. 2004;200:783–795. doi: 10.1084/jem.20040254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Inouye S, et al. Heat shock transcription factor 1 opens chromatin structure of interleukin-6 promoter to facilitate binding of an activator or a repressor. J Biol Chem. 2007;282:33210–33217. doi: 10.1074/jbc.M704471200. [DOI] [PubMed] [Google Scholar]
  • 167.House SD, et al. Effects of heat shock, stannous chloride, and gallium nitrate on the rat inflammatory response. Cell Stress Chaperones. 2001;6:164–171. doi: 10.1379/1466-1268(2001)006<0164:eohssc>2.0.co;2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ensor JE, et al. Differential effects of hyperthermia on macrophage interleukin-6 and tumor necrosis factor-alpha expression. Am J Physiol. 1994;266:C967–C974. doi: 10.1152/ajpcell.1994.266.4.C967. [DOI] [PubMed] [Google Scholar]
  • 169.Fairchild KD, Viscardi RM, Hester L, Singh IS, Hasday JD. Effects of hypothermia and hyperthermia on cytokine production by cultured human mononuclear phagocytes from adults and newborns. J Interferon Cytokine Res. 2000;20:1049–1055. doi: 10.1089/107999000750053708. [DOI] [PubMed] [Google Scholar]
  • 170.Hagiwara S, Iwasaka H, Matsumoto S, Noguchi T. Changes in cell culture temperature alter release of inflammatory mediators in murine macrophagic RAW264.7 cells. Inflamm Res. 2007;56:297–303. doi: 10.1007/s00011-007-6161-z. [DOI] [PubMed] [Google Scholar]
  • 171.Cooper ZA, et al. Febrile-range temperature modifies cytokine gene expression in LPS-stimulated macrophages by differentially modifying NF-{kappa}B recruitment to cytokine gene promoters. Am J Physiol Cell Physiol. 2010;298:C171–C181. doi: 10.1152/ajpcell.00346.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ensor JE, Crawford EK, Hasday JD. Warming macrophages to febrile range destabilizes tumor necrosis factor-alpha mRNA without inducing heat shock. Am J Physiol. 1995;269:C1140–C1146. doi: 10.1152/ajpcell.1995.269.5.C1140. [DOI] [PubMed] [Google Scholar]
  • 173.Singh IS, He JR, Calderwood S, Hasday JD. A high affinity HSF-1 binding site in the 5'-untranslated region of the murine tumor necrosis factor-alpha gene is a transcriptional repressor. J Biol Chem. 2002;277:4981–4988. doi: 10.1074/jbc.M108154200. [DOI] [PubMed] [Google Scholar]
  • 174.Fiuza C, et al. Inflammation-promoting activity of HMGB1 on human microvascular endothelial cells. Blood. 2003;101:2652–2660. doi: 10.1182/blood-2002-05-1300. [DOI] [PubMed] [Google Scholar]
  • 175.Lee CT, et al. Defining immunological impact and therapeutic benefit of mild heating in a murine model of arthritis. PLoS One. 2015;10:e0120327. doi: 10.1371/journal.pone.0120327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359. doi: 10.1152/physrev.00015.2003. [DOI] [PubMed] [Google Scholar]
  • 177.Cannon B, Nedergaard J. Nonshivering thermogenesis and its adequate measurement in metabolic studies. J Exp Biol. 2011;214:242–253. doi: 10.1242/jeb.050989. [DOI] [PubMed] [Google Scholar]
  • 178.Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve--an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev. 2000;52:595–638. [PubMed] [Google Scholar]
  • 179.Eng JW, et al. A nervous tumor microenvironment: the impact of adrenergic stress on cancer cells, immunosuppression, and immunotherapeutic response. Cancer Immunol Immunother. 2014;63:1115–1128. doi: 10.1007/s00262-014-1617-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Padgett DA, Glaser R. How stress influences the immune response. Trends Immunol. 2003;24:444–448. doi: 10.1016/s1471-4906(03)00173-x. [DOI] [PubMed] [Google Scholar]
  • 181.Nakai A, Hayano Y, Furuta F, Noda M, Suzuki K. Control of lymphocyte egress from lymph nodes through beta2-adrenergic receptors. J Exp Med. 2014;211:2583–2598. doi: 10.1084/jem.20141132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Slota C, Shi A, Chen G, Bevans M, Weng NP. Norepinephrine preferentially modulates memory CD8 T cell function inducing inflammatory cytokine production and reducing proliferation in response to activation. Brain Behav Immun. 2015 doi: 10.1016/j.bbi.2015.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Nguyen KD, et al. Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis. Nature. 2011;480:104–108. doi: 10.1038/nature10653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Kokolus KM, et al. Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature. Proc Natl Acad Sci U S A. 2013;110:20176–20181. doi: 10.1073/pnas.1304291110. References 183–184 establish that norepinephrine-driven thermogenesis shapes the phenotype of the immune response by altering macrophage differentiation and anti-tumor immunity.
  • 185.Kokolus KM, et al. Stressful presentations: mild cold stress in laboratory mice influences phenotype of dendritic cells in naive and tumor-bearing mice. Front Immunol. 2014;5:23. doi: 10.3389/fimmu.2014.00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Eng JW, et al. Housing temperature-induced stress drives therapeutic resistance in murine tumour models through beta2-adrenergic receptor activation. Nat Commun. 2015;6:6426. doi: 10.1038/ncomms7426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Glaser R, Kiecolt-Glaser JK. Stress-induced immune dysfunction: implications for health. Nat Rev Immunol. 2005;5:243–251. doi: 10.1038/nri1571. [DOI] [PubMed] [Google Scholar]
  • 188.Karp CL, Murray PJ. Non-canonical alternatives: what a macrophage is 4. J Exp Med. 2012;209:427–431. doi: 10.1084/jem.20120295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Di YP, Repasky EA, Subjeck JR. Distribution of HSP70, protein kinase C, and spectrin is altered in lymphocytes during a fever-like hyperthermia exposure. J Cell Physiol. 1997;172:44–54. doi: 10.1002/(SICI)1097-4652(199707)172:1<44::AID-JCP5>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 190.Tulapurkar ME, Asiegbu BE, Singh IS, Hasday JD. Hyperthermia in the febrile range induces HSP72 expression proportional to exposure temperature but not to HSF-1 DNA-binding activity in human lung epithelial A549 cells. Cell Stress Chaperones. 2009;14:499–508. doi: 10.1007/s12192-009-0103-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Gothard LQ, Ruffner ME, Woodward JG, Park-Sarge OK, Sarge KD. Lowered temperature set point for activation of the cellular stress response in T-lymphocytes. J Biol Chem. 2003;278:9322–9326. doi: 10.1074/jbc.M209412200. [DOI] [PubMed] [Google Scholar]
  • 192.Rossi A, Coccia M, Trotta E, Angelini M, Santoro MG. Regulation of cyclooxygenase-2 expression by heat: a novel aspect of heat shock factor 1 function in human cells. PLoS One. 2012;7:e31304. doi: 10.1371/journal.pone.0031304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Chatterjee M, et al. STAT3 and MAPK signaling maintain overexpression of heat shock proteins 90alpha and beta in multiple myeloma cells, which critically contribute to tumor-cell survival. Blood. 2007;109:720–728. doi: 10.1182/blood-2006-05-024372. [DOI] [PubMed] [Google Scholar]
  • 194.Marubayashi S, et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest. 2010;120:3578–3593. doi: 10.1172/JCI42442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sato N, et al. Involvement of heat-shock protein 90 in the interleukin-6-mediated signaling pathway through STAT3. Biochem Biophys Res Commun. 2003;300:847–852. doi: 10.1016/s0006-291x(02)02941-8. [DOI] [PubMed] [Google Scholar]
  • 196.Shang L, Tomasi TB. The heat shock protein 90-CDC37 chaperone complex is required for signaling by types I and II interferons. J Biol Chem. 2006;281:1876–1884. doi: 10.1074/jbc.M509901200. [DOI] [PubMed] [Google Scholar]
  • 197.Weigert O, et al. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition. J Exp Med. 2012;209:259–273. doi: 10.1084/jem.20111694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Patapoutian A, Peier AM, Story GM, Viswanath V. ThermoTRP channels and beyond: mechanisms of temperature sensation. Nat Rev Neurosci. 2003;4:529–539. doi: 10.1038/nrn1141. [DOI] [PubMed] [Google Scholar]
  • 199.Ramsey IS, Delling M, Clapham DE. An introduction to TRP channels. Annu Rev Physiol. 2006;68:619–647. doi: 10.1146/annurev.physiol.68.040204.100431. [DOI] [PubMed] [Google Scholar]
  • 200.Trepel J, Mollapour M, Giaccone G, Neckers L. Targeting the dynamic HSP90 complex in cancer. Nat Rev Cancer. 2010;10:537–549. doi: 10.1038/nrc2887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Zou J, Guo Y, Guettouche T, Smith DF, Voellmy R. Repression of heat shock transcription factor HSF1 activation by HSP90 (HSP90 complex) that forms a stress-sensitive complex with HSF1. Cell. 1998;94:471–480. doi: 10.1016/s0092-8674(00)81588-3. [DOI] [PubMed] [Google Scholar]
  • 202.Ahn SG, Thiele DJ. Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress. Genes Dev. 2003;17:516–528. doi: 10.1101/gad.1044503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer. 2005;5:761–772. doi: 10.1038/nrc1716. [DOI] [PubMed] [Google Scholar]
  • 204.Neckers L, Workman P. Hsp90 molecular chaperone inhibitors: are we there yet? Clin Cancer Res. 2012;18:64–76. doi: 10.1158/1078-0432.CCR-11-1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Muralidharan S, Mandrekar P. Cellular stress response and innate immune signaling: integrating pathways in host defense and inflammation. J Leukoc Biol. 2013;94:1167–1184. doi: 10.1189/jlb.0313153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. 2014;14:199–208. doi: 10.1038/nrc3672. [DOI] [PubMed] [Google Scholar]
  • 207.Repasky EA, Evans SS, Dewhirst MW. Temperature matters! And why it should matter to tumor immunologists. Cancer Immunol Res. 2013;1:210–216. doi: 10.1158/2326-6066.CIR-13-0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Haemmerich D, Laeseke PF. Thermal tumour ablation: devices, clinical applications and future directions. Int J Hyperthermia. 2005;21:755–760. doi: 10.1080/02656730500226423. [DOI] [PubMed] [Google Scholar]
  • 209.Issels RD, et al. Neo-adjuvant chemotherapy alone or with regional hyperthermia for localised high-risk soft-tissue sarcoma: a randomised phase 3 multicentre study. Lancet Oncol. 2010;11:561–570. doi: 10.1016/S1470-2045(10)70071-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Wessalowski R, et al. Regional deep hyperthermia for salvage treatment of children and adolescents with refractory or recurrent non-testicular malignant germ-cell tumours: an open-label, non-randomised, single-institution, phase 2 study. Lancet Oncol. 2013;14:843–852. doi: 10.1016/S1470-2045(13)70271-7. [DOI] [PubMed] [Google Scholar]
  • 211. Jones EL, et al. Randomized trial of hyperthermia and radiation for superficial tumors. J Clin Oncol. 2005;23:3079–3085. doi: 10.1200/JCO.2005.05.520. References 209–211 demonstrate that thermal therapy has a clinical benefit when used in an adjuvant setting in combination with chemotherapy or radiotherapy.
  • 212.Kong G, Braun RD, Dewhirst MW. Characterization of the effect of hyperthermia on nanoparticle extravasation from tumor vasculature. Cancer Res. 2001;61:3027–3032. [PubMed] [Google Scholar]
  • 213.Sen A, et al. Mild elevation of body temperature reduces tumor interstitial fluid pressure and hypoxia and enhances efficacy of radiotherapy in murine tumor models. Cancer Res. 2011;71:3872–3880. doi: 10.1158/0008-5472.CAN-10-4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Xu Y, et al. Fever-range whole body hyperthermia increases the number of perfused tumor blood vessels and therapeutic efficacy of liposomally encapsulated doxorubicin. Int J Hyperthermia. 2007;23:513–527. doi: 10.1080/02656730701666112. [DOI] [PubMed] [Google Scholar]
  • 215.Song CW, Park HJ, Lee CK, Griffin R. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia. 2005;21:761–767. doi: 10.1080/02656730500204487. [DOI] [PubMed] [Google Scholar]
  • 216.Dewhirst MW, Landon CD, Hofmann CL, Stauffer PR. Novel approaches to treatment of hepatocellular carcinoma and hepatic metastases using thermal ablation and thermosensitive liposomes. Surg Oncol Clin N Am. 2013;22:545–561. doi: 10.1016/j.soc.2013.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med. 2014;65:185–202. doi: 10.1146/annurev-med-092012-112807. [DOI] [PubMed] [Google Scholar]
  • 218.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12:269–281. doi: 10.1038/nri3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–277. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Wolchok JD, Chan TA. Cancer: Antitumour immunity gets a boost. Nature. 2014;515:496–498. doi: 10.1038/515496a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Guo J, et al. Intratumoral injection of dendritic cells in combination with local hyperthermia induces systemic antitumor effect in patients with advanced melanoma. Int J Cancer. 2007;120:2418–2425. doi: 10.1002/ijc.22551. [DOI] [PubMed] [Google Scholar]
  • 222.Mukhopadhaya A, et al. Localized hyperthermia combined with intratumoral dendritic cells induces systemic antitumor immunity. Cancer Res. 2007;67:7798–7806. doi: 10.1158/0008-5472.CAN-07-0203. [DOI] [PubMed] [Google Scholar]
  • 223.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES